1
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
2
|
Dabiri H, Mortezaei Z. Genome-wide association study of therapeutic response to statin drugs in cardiovascular disease. Sci Rep 2024; 14:18005. [PMID: 39097628 PMCID: PMC11297937 DOI: 10.1038/s41598-024-68665-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 07/26/2024] [Indexed: 08/05/2024] Open
Abstract
Cardiovascular disease (CVD) is one of the main causes of death in the world. The increased level of blood cholesterol is significantly correlated to CVD incidents. Statins are a group of drugs that decrease the synthesis of cholesterol in the liver by inhibiting the final enzyme of the pathway named HMG-CoA reductase. Several investigations showed that different patients give different responses to the administration of statin drugs according to their genetic background. In this research study, using Genome-Wide Association Studies (GWAS) data analysis methods, such as the SimpleM statistical approach and genomic connection matrix, we tried to discover the novel candidate SNPs that were involved in response to statin drugs. The investigation was carried out using 3,221 cardiovascular patients' data about genotypes and phenotypes of two important parameters including total cholesterol, and LDL level, in response to statin administration. Functional annotation of nearest genes to candidate SNPs was also carried out by using comprehensive databases and tools such as BioMart-Ensembl, UCSC, NCBI, and WebGestalt software. Our results represented eight novel SNPs (rs10820084, rs4803750, rs10989887, rs1966503, rs17502794, rs10785232, rs484071, rs4785621) significantly associated with statin response in different individual cardiovascular patients for the first time. In addition, the groups of genes that are close to the SNPs were also represented and evaluated in detail. Our results illustrated that some of the genes such as BAAT, BCL3, and CMTM6 have a direct functional impact on cholesterol level or LDL biosynthesis which confirmed the effects of neighbor SNPs on the response to statin drugs. Today, finding the loci, genes, and molecular mechanisms involved in the response to drugs is of great importance in pharmacogenomics and personalized medicine.
Collapse
Affiliation(s)
- Hamed Dabiri
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Mortezaei
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
4
|
Bueno Marinas M, Cason M, Bariani R, Celeghin R, De Gaspari M, Pinci S, Cipriani A, Rigato I, Zorzi A, Rizzo S, Thiene G, Perazzolo Marra M, Corrado D, Basso C, Bauce B, Pilichou K. A Comprehensive Analysis of Non-Desmosomal Rare Genetic Variants in Arrhythmogenic Cardiomyopathy: Integrating in Padua Cohort Literature-Derived Data. Int J Mol Sci 2024; 25:6267. [PMID: 38892455 PMCID: PMC11173278 DOI: 10.3390/ijms25116267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited myocardial disease at risk of sudden death. Genetic testing impacts greatly in ACM diagnosis, but gene-disease associations have yet to be determined for the increasing number of genes included in clinical panels. Genetic variants evaluation was undertaken for the most relevant non-desmosomal disease genes. We retrospectively studied 320 unrelated Italian ACM patients, including 243 cases with predominant right-ventricular (ARVC) and 77 cases with predominant left-ventricular (ALVC) involvement, who did not carry pathogenic/likely pathogenic (P/LP) variants in desmosome-coding genes. The aim was to assess rare genetic variants in transmembrane protein 43 (TMEM43), desmin (DES), phospholamban (PLN), filamin c (FLNC), cadherin 2 (CDH2), and tight junction protein 1 (TJP1), based on current adjudication guidelines and reappraisal on reported literature data. Thirty-five rare genetic variants, including 23 (64%) P/LP, were identified in 39 patients (16/243 ARVC; 23/77 ALVC): 22 FLNC, 9 DES, 2 TMEM43, and 2 CDH2. No P/LP variants were found in PLN and TJP1 genes. Gene-based burden analysis, including P/LP variants reported in literature, showed significant enrichment for TMEM43 (3.79-fold), DES (10.31-fold), PLN (117.8-fold) and FLNC (107-fold). A non-desmosomal rare genetic variant is found in a minority of ARVC patients but in about one third of ALVC patients; as such, clinical decision-making should be driven by genes with robust evidence. More than two thirds of non-desmosomal P/LP variants occur in FLNC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy; (M.B.M.); (M.C.); (R.B.); (R.C.); (M.D.G.); (S.P.); (A.C.); (I.R.); (A.Z.); (S.R.); (G.T.); (M.P.M.); (D.C.); (B.B.); (K.P.)
| | | | | |
Collapse
|
5
|
Monte E, Furihata T, Wang G, Perea-Gil I, Wei E, Chaib H, Nair R, Guevara JV, Mares R, Cheng X, Zhuge Y, Black K, Serrano R, Dagan-Rosenfeld O, Maguire P, Mercola M, Karakikes I, Wu JC, Snyder MP. Personalized transcriptome signatures in a cardiomyopathy stem cell biobank. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593618. [PMID: 38798547 PMCID: PMC11118309 DOI: 10.1101/2024.05.10.593618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND There is growing evidence that pathogenic mutations do not fully explain hypertrophic (HCM) or dilated (DCM) cardiomyopathy phenotypes. We hypothesized that if a patient's genetic background was influencing cardiomyopathy this should be detectable as signatures in gene expression. We built a cardiomyopathy biobank resource for interrogating personalized genotype phenotype relationships in human cell lines. METHODS We recruited 308 diseased and control patients for our cardiomyopathy stem cell biobank. We successfully reprogrammed PBMCs (peripheral blood mononuclear cells) into induced pluripotent stem cells (iPSCs) for 300 donors. These iPSCs underwent whole genome sequencing and were differentiated into cardiomyocytes for RNA-seq. In addition to annotating pathogenic variants, mutation burden in a panel of cardiomyopathy genes was assessed for correlation with echocardiogram measurements. Line-specific co-expression networks were inferred to evaluate transcriptomic subtypes. Drug treatment targeted the sarcomere, either by activation with omecamtiv mecarbil or inhibition with mavacamten, to alter contractility. RESULTS We generated an iPSC biobank from 300 donors, which included 101 individuals with HCM and 88 with DCM. Whole genome sequencing of 299 iPSC lines identified 78 unique pathogenic or likely pathogenic mutations in the diseased lines. Notably, only DCM lines lacking a known pathogenic or likely pathogenic mutation replicated a finding in the literature for greater nonsynonymous SNV mutation burden in 102 cardiomyopathy genes to correlate with lower left ventricular ejection fraction in DCM. We analyzed RNA-sequencing data from iPSC-derived cardiomyocytes for 102 donors. Inferred personalized co-expression networks revealed two transcriptional subtypes of HCM. The first subtype exhibited concerted activation of the co-expression network, with the degree of activation reflective of the disease severity of the donor. In contrast, the second HCM subtype and the entire DCM cohort exhibited partial activation of the respective disease network, with the strength of specific gene by gene relationships dependent on the iPSC-derived cardiomyocyte line. ADCY5 was the largest hubnode in both the HCM and DCM networks and partially corrected in response to drug treatment. CONCLUSIONS We have a established a stem cell biobank for studying cardiomyopathy. Our analysis supports the hypothesis the genetic background influences pathologic gene expression programs and support a role for ADCY5 in cardiomyopathy.
Collapse
Affiliation(s)
- Emma Monte
- Department of Genetics, Stanford University School of Medicine
| | | | - Guangwen Wang
- Department of Genetics, Stanford University School of Medicine
| | - Isaac Perea-Gil
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Cardiothoracic Surgery, Stanford University School of Medicine
| | - Eric Wei
- Department of Genetics, Stanford University School of Medicine
| | - Hassan Chaib
- Department of Genetics, Stanford University School of Medicine
| | - Ramesh Nair
- Department of Genetics, Stanford University School of Medicine
| | - Julio Vicente Guevara
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Rene Mares
- Department of Genetics, Stanford University School of Medicine
| | - Xun Cheng
- Department of Genetics, Stanford University School of Medicine
| | - Yan Zhuge
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Katelyn Black
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Ricardo Serrano
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | | | - Peter Maguire
- Department of Genetics, Stanford University School of Medicine
| | - Mark Mercola
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Ioannis Karakikes
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Cardiothoracic Surgery, Stanford University School of Medicine
| | - Joseph C Wu
- Cardiovascular Institute, Stanford University School of Medicine
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine
| | | |
Collapse
|
6
|
Chua CJ, Morrissette-McAlmon J, Tung L, Boheler KR. Understanding Arrhythmogenic Cardiomyopathy: Advances through the Use of Human Pluripotent Stem Cell Models. Genes (Basel) 2023; 14:1864. [PMID: 37895213 PMCID: PMC10606441 DOI: 10.3390/genes14101864] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiomyopathies (CMPs) represent a significant healthcare burden and are a major cause of heart failure leading to premature death. Several CMPs are now recognized to have a strong genetic basis, including arrhythmogenic cardiomyopathy (ACM), which predisposes patients to arrhythmic episodes. Variants in one of the five genes (PKP2, JUP, DSC2, DSG2, and DSP) encoding proteins of the desmosome are known to cause a subset of ACM, which we classify as desmosome-related ACM (dACM). Phenotypically, this disease may lead to sudden cardiac death in young athletes and, during late stages, is often accompanied by myocardial fibrofatty infiltrates. While the pathogenicity of the desmosome genes has been well established through animal studies and limited supplies of primary human cells, these systems have drawbacks that limit their utility and relevance to understanding human disease. Human induced pluripotent stem cells (hiPSCs) have emerged as a powerful tool for modeling ACM in vitro that can overcome these challenges, as they represent a reproducible and scalable source of cardiomyocytes (CMs) that recapitulate patient phenotypes. In this review, we provide an overview of dACM, summarize findings in other model systems linking desmosome proteins with this disease, and provide an up-to-date summary of the work that has been conducted in hiPSC-cardiomyocyte (hiPSC-CM) models of dACM. In the context of the hiPSC-CM model system, we highlight novel findings that have contributed to our understanding of disease and enumerate the limitations, prospects, and directions for research to consider towards future progress.
Collapse
Affiliation(s)
- Christianne J. Chua
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Graziano F, Cipriani A, Balla D, Bondarev S, Marra MP, Bauce B, Vágó H, Corrado D, Zorzi A. Evolving spectrum of arrhythmogenic cardiomyopathy: Implications for Sports Cardiology. Clin Cardiol 2023; 46:1072-1081. [PMID: 37357443 PMCID: PMC10540004 DOI: 10.1002/clc.24069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/27/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic heart muscle disease, structurally characterized by progressive fibro-fatty replacement of the normal myocardium and clinically by ventricular arrhythmias (VAs). Predominantly thanks to the use of cardiac magnetic resonance, we have learnt that the spectrum of the disease encompasses not only the classical right ventricular phenotype, but also biventricular and left dominant variants. Sport activity contributes to the phenotypic expression and progression of ACM and may trigger life-threatening VAs and sudden cardiac death (SCD). We conducted a review of the literature about ACM and its implications in Sport Cardiology and summarized the main findings in this topic. Early identification of affected athletes through preparticipation screening (PPS) is fundamental but, while classical right-ventricular or biventricular phenotypes are usually suspected because of electrocardiogram (ECG) and echocardiographic abnormalities, variants with predominant left ventricular involvement are often characterized by normal ECG and unremarkable echocardiography. Usually the only manifestations of such variants are exercise-induced VAs and for this reason exercise testing may empower the diagnostic yield of the PPS. Patients with ACM are not eligible to competitive sports activity, but low-to-moderate intensity physical activity under medical supervision is possible in most cases.
Collapse
Affiliation(s)
- Francesca Graziano
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Alberto Cipriani
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Dorottya Balla
- Department of Sports MedicineSemmelweis UniversityBudapestHungary
| | - Sergei Bondarev
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Martina Perazzolo Marra
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Barbara Bauce
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Hajnalka Vágó
- Department of Sports MedicineSemmelweis UniversityBudapestHungary
- Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Domenico Corrado
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| | - Alessandro Zorzi
- Department of Cardiac, Thoracic and Vascular Sciences and Public HealthUniversity of PaduaPadovaItaly
| |
Collapse
|
8
|
van Ham WB, Meijboom EEM, Ligtermoet ML, Nikkels PGJ, van Veen TAB. Maturation and Function of the Intercalated Disc: Report of Two Pediatric Cases Focusing on Cardiac Development and Myocardial Hyperplasia. J Cardiovasc Dev Dis 2023; 10:354. [PMID: 37623366 PMCID: PMC10455643 DOI: 10.3390/jcdd10080354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
The development of the normal human heart, ranging from gestational age to the mature adult heart, relies on a very delicate and timely orchestrated order of processes. One of the most striking alterations in time is the gradual extinction of the ability for cardiomyocytes to proliferate. Once passing this event, cardiomyocytes grow and increase in contractile strength by means of physiological hypertrophy. This process, importantly, seems to depend on an adequate development of electromechanical coupling that is achieved by the appropriate formation of the intercellular junction named the intercalated disc (ICD). In this report, we describe two sudden death cases of young and apparently healthy-born individuals without external abnormalities compared to an age-matched control. Histological examination, including the comparison with the age-matched and histology-matched controls, showed a disturbed formation of the protein machinery composing the electromechanical junctions at the ICD and an increased nuclei count for both patients. As a cause or consequence, cardiomyocytes in both sudden death cases showed signs of a delayed developmental stage, presumably resulting in an exaggerated degree of hyperplasia.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Esmeralda E. M. Meijboom
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Merel L. Ligtermoet
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Peter G. J. Nikkels
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
9
|
Thiene G, Basso C, Pilichou K, Bueno Marinas M. Desmosomal Arrhythmogenic Cardiomyopathy: The Story Telling of a Genetically Determined Heart Muscle Disease. Biomedicines 2023; 11:2018. [PMID: 37509658 PMCID: PMC10377062 DOI: 10.3390/biomedicines11072018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The history of arrhythmogenic cardiomyopathy (AC) as a genetically determined desmosomal disease started since the original discovery by Lancisi in a four-generation family, published in 1728. Contemporary history at the University of Padua started with Dalla Volta, who haemodynamically investigated patients with "auricularization" of the right ventricle, and with Nava, who confirmed familiarity. The contemporary knowledge advances consisted of (a) AC as a heart muscle disease with peculiar electrical instability of the right ventricle; (b) the finding of pathological substrates, in keeping with a myocardial dystrophy; (c) the inclusion of AC in the cardiomyopathies classification; (d) AC as the main cause of sudden death in athletes; (e) the discovery of the culprit genes coding proteins of the intercalated disc (desmosome); (f) progression in clinical diagnosis with specific ECG abnormalities, angiocardiography, endomyocardial biopsy, 2D echocardiography, electron anatomic mapping and cardiac magnetic resonance; (g) the discovery of left ventricular AC; (h) prevention of SCD with the invention and application of the lifesaving implantable cardioverter defibrillator and external defibrillator scattered in public places and playgrounds as well as the ineligibility for competitive sport activity for AC patients; (i) genetic screening of the proband family to unmask asymptomatic carriers. Nondesmosomal ACs, with a phenotype overlapping desmosomal AC, are also treated, including genetics: Transmembrane protein 43, SCN5A, Desmin, Phospholamban, Lamin A/C, Filamin C, Cadherin 2, Tight junction protein 1.
Collapse
Affiliation(s)
- Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Maria Bueno Marinas
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| |
Collapse
|
10
|
Yeruva S, Stangner K, Jungwirth A, Hiermaier M, Shoykhet M, Kugelmann D, Hertl M, Egami S, Ishii N, Koga H, Hashimoto T, Weis M, Beckmann BM, Biller R, Schüttler D, Kääb S, Waschke J. Catalytic antibodies in arrhythmogenic cardiomyopathy patients cleave desmoglein 2 and N-cadherin and impair cardiomyocyte cohesion. Cell Mol Life Sci 2023; 80:203. [PMID: 37450050 PMCID: PMC10348947 DOI: 10.1007/s00018-023-04853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
AIMS Arrhythmogenic cardiomyopathy (AC) is a severe heart disease predisposing to ventricular arrhythmias and sudden cardiac death caused by mutations affecting intercalated disc (ICD) proteins and aggravated by physical exercise. Recently, autoantibodies targeting ICD proteins, including the desmosomal cadherin desmoglein 2 (DSG2), were reported in AC patients and were considered relevant for disease development and progression, particularly in patients without underlying pathogenic mutations. However, it is unclear at present whether these autoantibodies are pathogenic and by which mechanisms show specificity for DSG2 and thus can be used as a diagnostic tool. METHODS AND RESULTS IgG fractions were purified from 15 AC patients and 4 healthy controls. Immunostainings dissociation assays, atomic force microscopy (AFM), Western blot analysis and Triton X-100 assays were performed utilizing human heart left ventricle tissue, HL-1 cells and murine cardiac slices. Immunostainings revealed that autoantibodies against ICD proteins are prevalent in AC and most autoantibody fractions have catalytic properties and cleave the ICD adhesion molecules DSG2 and N-cadherin, thereby reducing cadherin interactions as revealed by AFM. Furthermore, most of the AC-IgG fractions causing loss of cardiomyocyte cohesion activated p38MAPK, which is known to contribute to a loss of desmosomal adhesion in different cell types, including cardiomyocytes. In addition, p38MAPK inhibition rescued the loss of cardiomyocyte cohesion induced by AC-IgGs. CONCLUSION Our study demonstrates that catalytic autoantibodies play a pathogenic role by cleaving ICD cadherins and thereby reducing cardiomyocyte cohesion by a mechanism involving p38MAPK activation. Finally, we conclude that DSG2 cleavage by autoantibodies could be used as a diagnostic tool for AC.
Collapse
Affiliation(s)
- Sunil Yeruva
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Konstanze Stangner
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Anna Jungwirth
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Maria Shoykhet
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
- Department of Otorhinolarynology, Technical University of Munich and University Hospital rechts der Isar, Ismaningerstrasse 22, 81675, Munich, Germany
| | - Daniela Kugelmann
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps-University Marburg, 35037, Marburg, Germany
| | - Shohei Egami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Norito Ishii
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Hiroshi Koga
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Hashimoto
- Department of Dermatology, Graduate School of Medicine, Osaka City Metropolitan University, Osaka, Japan
| | - Michael Weis
- Krankenhaus Neuwittelsbach, Fachklinik Für Innere Medizin, Munich, Germany
| | - Britt-Maria Beckmann
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians University Munich (LMU), Campus Großhadern, Munich, Germany
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Ruth Biller
- ARVC-Selbsthilfe E.V, Patient Association, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians University Munich (LMU), Campus Großhadern, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians University Munich (LMU), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICON), LMU Munich, Munich, Germany
| | - Stefan Kääb
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians University Munich (LMU), Campus Großhadern, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance (MHA), Partner Site Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICON), LMU Munich, Munich, Germany
- Member of the European Reference Network for rare, low prevalance and complex diseases of the heart , ERN GUARD-Heart, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstrasse 11, 80336, Munich, Germany.
| |
Collapse
|
11
|
Bueno-Beti C, Asimaki A. Cheek-Pro-Heart: What Can the Buccal Mucosa Do for Arrhythmogenic Cardiomyopathy? Biomedicines 2023; 11:biomedicines11041207. [PMID: 37189825 DOI: 10.3390/biomedicines11041207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heart muscle disease associated with ventricular arrhythmias and a high risk of sudden cardiac death (SCD). Although the disease was described over 40 years ago, its diagnosis is still difficult. Several studies have identified a set of five proteins (plakoglobin, Cx43, Nav1.5, SAP97 and GSK3β), which are consistently re-distributed in myocardial samples from ACM patients. Not all protein shifts are specific to ACM, but their combination has provided us with a molecular signature for the disease, which has greatly aided post-mortem diagnosis of SCD victims. The use of this signature, however, was heretofore restricted in living patients, as the analysis requires a heart sample. Recent studies have shown that buccal cells behave similarly to the heart in terms of protein re-localization. Protein shifts are associated with disease onset, deterioration and favorable response to anti-arrhythmic therapy. Accordingly, buccal cells can be used as a surrogate for the myocardium to aid diagnosis, risk stratification and even monitor response to pharmaceutical interventions. Buccal cells can also be kept in culture, hence providing an ex vivo model from the patient, which can offer insights into the mechanisms of disease pathogenesis, including drug response. This review summarizes how the cheek can aid the heart in the battle against ACM.
Collapse
Affiliation(s)
- Carlos Bueno-Beti
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW17 0RE, UK
| |
Collapse
|
12
|
Tsui H, van Kampen SJ, Han SJ, Meraviglia V, van Ham WB, Casini S, van der Kraak P, Vink A, Yin X, Mayr M, Bossu A, Marchal GA, Monshouwer-Kloots J, Eding J, Versteeg D, de Ruiter H, Bezstarosti K, Groeneweg J, Klaasen SJ, van Laake LW, Demmers JAA, Kops GJPL, Mummery CL, van Veen TAB, Remme CA, Bellin M, van Rooij E. Desmosomal protein degradation as an underlying cause of arrhythmogenic cardiomyopathy. Sci Transl Med 2023; 15:eadd4248. [PMID: 36947592 DOI: 10.1126/scitranslmed.add4248] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 03/01/2023] [Indexed: 03/24/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive cardiac disease. Many patients with ACM harbor mutations in desmosomal genes, predominantly in plakophilin-2 (PKP2). Although the genetic basis of ACM is well characterized, the underlying disease-driving mechanisms remain unresolved. Explanted hearts from patients with ACM had less PKP2 compared with healthy hearts, which correlated with reduced expression of desmosomal and adherens junction (AJ) proteins. These proteins were also disorganized in areas of fibrotic remodeling. In vitro data from human-induced pluripotent stem cell-derived cardiomyocytes and microtissues carrying the heterozygous PKP2 c.2013delC pathogenic mutation also displayed impaired contractility. Knockin mice carrying the equivalent heterozygous Pkp2 c.1755delA mutation recapitulated changes in desmosomal and AJ proteins and displayed cardiac dysfunction and fibrosis with age. Global proteomics analysis of 4-month-old heterozygous Pkp2 c.1755delA hearts indicated involvement of the ubiquitin-proteasome system (UPS) in ACM pathogenesis. Inhibition of the UPS in mutant mice increased area composita proteins and improved calcium dynamics in isolated cardiomyocytes. Additional proteomics analyses identified lysine ubiquitination sites on the desmosomal proteins, which were more ubiquitinated in mutant mice. In summary, we show that a plakophilin-2 mutation can lead to decreased desmosomal and AJ protein expression through a UPS-dependent mechanism, which preceded cardiac remodeling. These findings suggest that targeting protein degradation and improving desmosomal protein stability may be a potential therapeutic strategy for the treatment of ACM.
Collapse
Affiliation(s)
- Hoyee Tsui
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Sebastiaan Johannes van Kampen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Su Ji Han
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, University Medical Center, Leiden, 2333 ZA, Netherlands
| | - Willem B van Ham
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CM, Netherlands
| | - Simona Casini
- Department of Clinical and Experimental Cardiology, University Medical Center Amsterdam, 1105 AZ, Netherlands
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, 3584 CX, Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, 3584 CX, Netherlands
| | - Xiaoke Yin
- James Black Centre, King's College, University of London, WC2R 2LS London, UK
| | - Manuel Mayr
- James Black Centre, King's College, University of London, WC2R 2LS London, UK
| | - Alexandre Bossu
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CM, Netherlands
| | - Gerard A Marchal
- Department of Clinical and Experimental Cardiology, University Medical Center Amsterdam, 1105 AZ, Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Joep Eding
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Hesther de Ruiter
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Erasmus Medical Center Rotterdam, 3015 CN, Netherlands
| | - Judith Groeneweg
- Department of Cardiology, University Medical Center Utrecht, 3584 CX, Netherlands
| | - Sjoerd J Klaasen
- Oncode Institute, Hubrecht Institute, Royal Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Linda W van Laake
- Department of Cardiology, University Medical Center Utrecht, 3584 CX, Netherlands
| | - Jeroen A A Demmers
- Proteomics Center, Erasmus Medical Center Rotterdam, 3015 CN, Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute, Royal Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, University Medical Center, Leiden, 2333 ZA, Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CM, Netherlands
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, University Medical Center Amsterdam, 1105 AZ, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, University Medical Center, Leiden, 2333 ZA, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT, Netherlands
- Department of Cardiology, University Medical Center Utrecht, 3584 CX, Netherlands
| |
Collapse
|
13
|
Li K, Jiang Y, Zeng Y, Zhou Y. Advances in Ion Channel, Non-Desmosomal Variants and Autophagic Mechanisms Implicated in Arrhythmogenic Cardiomyopathy. Curr Issues Mol Biol 2023; 45:2186-2200. [PMID: 36975511 PMCID: PMC10047275 DOI: 10.3390/cimb45030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heterogeneous disorder characterized by the replacement of cardiac myocytes with fibro-fatty tissues, leading to abnormal excitation-contraction (EC) coupling and a range of malignant events, such as ventricular tachycardia (VT), sudden cardiac death/arrest (SCD/A) and heart failure (HF). The concept of ACM has recently been ex-tended to include right ventricular cardiomyopathy (ARVC), left ventricular cardiomyopathy (ALVC) and biventricular cardiomyopathy. ARVC is generally seen as the most common type of ACM. The pathogenesis of ACM involves mutation variants in desmosomal or non-desmosomal gene loci, as well as various external factors, such as intense exercise, stress and infections. Ion channel alterations, autophagy and non-desmosomal variants are also important components in the development of ACM. As clinical practice enters the era of precision therapy, it is important to review recent studies on these topics to better diagnose and treat the molecular phase of ACM.
Collapse
Affiliation(s)
- Kexin Li
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yiyao Zeng
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
- Correspondence: ; Tel.: +86-512-65955026
| |
Collapse
|
14
|
Wang Y, Dobreva G. Epigenetics in LMNA-Related Cardiomyopathy. Cells 2023; 12:cells12050783. [PMID: 36899919 PMCID: PMC10001118 DOI: 10.3390/cells12050783] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Mutations in the gene for lamin A/C (LMNA) cause a diverse range of diseases known as laminopathies. LMNA-related cardiomyopathy is a common inherited heart disease and is highly penetrant with a poor prognosis. In the past years, numerous investigations using mouse models, stem cell technologies, and patient samples have characterized the phenotypic diversity caused by specific LMNA variants and contributed to understanding the molecular mechanisms underlying the pathogenesis of heart disease. As a component of the nuclear envelope, LMNA regulates nuclear mechanostability and function, chromatin organization, and gene transcription. This review will focus on the different cardiomyopathies caused by LMNA mutations, address the role of LMNA in chromatin organization and gene regulation, and discuss how these processes go awry in heart disease.
Collapse
Affiliation(s)
- Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), 68167 Mannheim, Germany
- Correspondence: (Y.W.); (G.D.)
| |
Collapse
|
15
|
Alblaihed L, Kositz C, Brady WJ, Al-Salamah T, Mattu A. Diagnosis and management of arrhythmogenic right ventricular cardiomyopathy. Am J Emerg Med 2023; 65:146-153. [PMID: 36638611 DOI: 10.1016/j.ajem.2022.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a genetic disorder of the myocardium that can lead to ventricular arrhythmia and sudden cardiac death. The condition has been identified as a significant cause of arrhythmic death among young people and athletes, therefore, early recognition of the disease by emergency clinicians is critical to prevent subsequent death. The diagnosis of ARVC can be very challenging and requires a systematic approach. This publication reviews the pathophysiology, classification, clinical presentations, and appropriate approach to diagnosis and management of ARVC.
Collapse
Affiliation(s)
- Leen Alblaihed
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S Paca Street, 6(th) Floor, Suite 200, Baltimore, MD 21201, United States of America.
| | - Christine Kositz
- Depratment of Emergency Medicine, University of Maryland Shore Medical Center at Easton, 219 S Washington St, Easton, MD 21601, United States of America
| | - William J Brady
- Department of Emergency Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Tareq Al-Salamah
- Department of Emergency Medicine, College of Medicine, King Saud University, PO Box 7805, Riyadh 11472, Saudi Arabia
| | - Amal Mattu
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S Paca Street, 6(th) Floor, Suite 200, Baltimore, MD 21201, United States of America
| |
Collapse
|
16
|
Using Zebrafish Animal Model to Study the Genetic Underpinning and Mechanism of Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24044106. [PMID: 36835518 PMCID: PMC9966228 DOI: 10.3390/ijms24044106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is largely an autosomal dominant genetic disorder manifesting fibrofatty infiltration and ventricular arrhythmia with predominantly right ventricular involvement. ACM is one of the major conditions associated with an increased risk of sudden cardiac death, most notably in young individuals and athletes. ACM has strong genetic determinants, and genetic variants in more than 25 genes have been identified to be associated with ACM, accounting for approximately 60% of ACM cases. Genetic studies of ACM in vertebrate animal models such as zebrafish (Danio rerio), which are highly amenable to large-scale genetic and drug screenings, offer unique opportunities to identify and functionally assess new genetic variants associated with ACM and to dissect the underlying molecular and cellular mechanisms at the whole-organism level. Here, we summarize key genes implicated in ACM. We discuss the use of zebrafish models, categorized according to gene manipulation approaches, such as gene knockdown, gene knock-out, transgenic overexpression, and CRISPR/Cas9-mediated knock-in, to study the genetic underpinning and mechanism of ACM. Information gained from genetic and pharmacogenomic studies in such animal models can not only increase our understanding of the pathophysiology of disease progression, but also guide disease diagnosis, prognosis, and the development of innovative therapeutic strategies.
Collapse
|
17
|
Castelletti S, Orini M, Vischer AS, McKenna WJ, Lambiase PD, Pantazis A, Crotti L. Circadian and Seasonal Pattern of Arrhythmic Events in Arrhythmogenic Cardiomyopathy Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2872. [PMID: 36833593 PMCID: PMC9956986 DOI: 10.3390/ijerph20042872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/22/2023] [Accepted: 01/28/2023] [Indexed: 05/28/2023]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease associated with an increased risk of life-threatening arrhythmias. The aim of the present study was to evaluate the association of ventricular arrhythmias (VA) with circadian and seasonal variation in ARVC. One hundred two ARVC patients with an implantable cardioverter defibrillator (ICD) were enrolled in the study. Arrhythmic events included (a) any initial ventricular tachycardia (VT) or fibrillation (VF) prompting ICD implantation, (b) any VT or non-sustained VT (NSVT) recorded by the ICD, and (c) appropriate ICD shocks/therapy. Differences in the annual incidence of events across seasons (winter, spring, summer, autumn) and period of the day (night, morning, afternoon, evening) were assessed both for all cardiac events and major arrhythmic events. In total, 67 events prior to implantation and 263 ICD events were recorded. These included 135 major (58 ICD therapies, 57 self-terminating VT, 20 sustained VT) and 148 minor (NSVT) events. A significant increase in the frequency of events was observed in the afternoon versus in the nights and mornings (p = 0.016). The lowest number of events was registered in the summer, with a peak in the winter (p < 0.001). Results were also confirmed when excluding NSVT. Arrhythmic events in ARVC follow a seasonal variation and a circadian rhythm. They are more prevalent in the late afternoon, the most active period of the day, and in the winter, supporting the role of physical activity and inflammation as triggers of events.
Collapse
Affiliation(s)
- Silvia Castelletti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiology, Piazzale Brescia 20, 20149 Milan, Italy
| | - Michele Orini
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Annina S. Vischer
- Medical Outpatient Department, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| | - William J. McKenna
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, University of A Coruña, 15001 A Coruña, Spain
| | - Pier D. Lambiase
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- The Barts Heart Centre, Barts Health NHS Trust, London E1 1BB, UK
| | - Antonios Pantazis
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
- Cardiovascular Research Centre, Royal Brompton and Harefield Hospitals, London SW3 6NP, UK
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiology, Piazzale Brescia 20, 20149 Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
18
|
Wen H, Chen W, Chen Y, Wei G, Ni T. Integrative analysis of Iso-Seq and RNA-seq reveals dynamic changes of alternative promoter, alternative splicing and alternative polyadenylation during Angiotensin II-induced senescence in rat primary aortic endothelial cells. Front Genet 2023; 14:1064624. [PMID: 36741323 PMCID: PMC9892061 DOI: 10.3389/fgene.2023.1064624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Abstract
In eukaryotes, alternative promoter (AP), alternative splicing (AS), and alternative polyadenylation (APA) are three crucial regulatory mechanisms that modulate message RNA (mRNA) diversity. Although AP, AS and APA are involved in diverse biological processess, whether they have dynamic changes in Angiotensin II (Ang II) induced senescence in rat primary aortic endothelial cells (RAECs), an important cellular model for studying cardiovascular disease, remains unclear. Here we integrated both PacBio single-molecule long-read isoform sequencing (Iso-Seq) and Illumina short-read RNA sequencing (RNA-seq) to analyze the changes of AP, AS and APA in Ang II-induced senescent RAECs. Iso-Seq generated 36,278 isoforms from 10,145 gene loci and 65.81% of these isoforms are novel, which were further cross-validated by public data obtained by other techonologies such as CAGE, PolyA-Seq and 3'READS. APA contributed most to novel isoforms, followed by AS and AP. Further investigation showed that AP, AS and APA could all contribute to the regulation of isoform, but AS has more dynamic changes compared to AP and APA upon Ang II stimulation. Genes undergoing AP, AS and APA in Ang II-treated cells are enriched in various pathways related to aging or senescence, suggesting that these molecular changes are involved in functional alterations during Ang II-induced senescence. Together, the present study largely improved the annotation of rat genome and revealed gene expression changes at isoform level, extending the understanding of the complexity of gene regulation in Ang II-treated RAECs, and also provided novel clues for discovering the regulatory mechanism undelying Ang II caused vascular senescence and diseases.
Collapse
Affiliation(s)
- Haimei Wen
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Chen
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yu Chen
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Gang Wei
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Ting Ni
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Chang E, Daboul R, Hanan A, Abdo M, Poulik J, Eskarous H, Hadjilambris AM, Shehata BM. Revisiting the Newly Modified Criteria for Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia (ARVC/D) and Reporting Newly Identified Genes. Fetal Pediatr Pathol 2022; 41:909-918. [PMID: 34854351 DOI: 10.1080/15513815.2021.2008564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Background: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia is an inherited cardiomyopathy, characterized by replacement of the RV muscle wall with fibrofatty tissue. The diagnosis is challenging, due to the absence of a unique presentation and a lack of specific reproducible diagnostic criteria. Materials and methods: Slides and additional clinical information including follow up from 16 cases were reviewed. Pediatric criteria of >30% of muscle replacement was used, instead of >40% as used in adults. Results: All 16 cases were confirmed by genetic testing and show ARVC/D. Applying the adult criteria, 7 cases would not have been categorized as ARVC/D. Conclusion: The modified pediatric criteria for ARVC/D should be used for pediatric patients. Better detection will aid in genetic counseling in order to identify those additional family members susceptible to sudden cardiac deaths so they can be followed optimally.
Collapse
Affiliation(s)
- Eric Chang
- Children's Hospital of Michigan, Detroit, MI, USA
| | - Rania Daboul
- Children's Hospital of Michigan, Detroit, MI, USA
| | - Abdul Hanan
- Children's Hospital of Michigan, Detroit, MI, USA
| | - Mena Abdo
- Children's Hospital of Michigan, Detroit, MI, USA
| | - Janet Poulik
- Children's Hospital of Michigan, Detroit, MI, USA
| | | | | | | |
Collapse
|
20
|
Spracklen TF, Keavney B, Laing N, Ntusi N, Shaboodien G. Modern genomic techniques in the identification of genetic causes of cardiomyopathy. Heart 2022; 108:1843-1850. [PMID: 35140110 DOI: 10.1136/heartjnl-2021-320424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/18/2022] [Indexed: 11/04/2022] Open
Abstract
Over the past three decades numerous disease-causing genes have been linked to the pathogenesis of heritable cardiomyopathies, but many causal genes are yet to be identified. Next-generation sequencing (NGS) platforms have revolutionised clinical testing capacity in familial cardiomyopathy. In this review, we summarise how NGS technologies have advanced our understanding of genetic non-syndromic cardiomyopathy over the last decade. First, 26 putative new disease-causing genes have been identified to date, mostly from whole-exome sequencing, and some of which (FLNC, MTO1, HCN4) have had a considerable clinical impact and are now included in routine diagnostic gene panels. Second, we consider challenges in variant interpretation and the importance of large-scale NGS population control cohorts for this purpose. Third, an emerging role of common variation in some forms of genetic cardiomyopathy is being elucidated through recent studies which have illustrated an additive effect of numerous polymorphic loci on cardiac parameters; this may explain phenotypic variability and low rates of genetic diagnosis from sequencing studies. Finally, we discuss the clinical utility of genetic testing in cardiomyopathy in Western settings, where NGS panel testing of core disease genes is currently recommended with possible implications for patient management. Given the findings of recent studies, whole-exome or whole-genome sequencing should be considered in patients of non-European ancestry with clearly familial disease, or severe paediatric disease, when no result is obtained on panel sequencing. The clinical utility of polygenic risk assessment needs to be investigated further in patients with unexplained dilated cardiomyopathy and hypertrophic cardiomyopathy in whom a pathogenic variant is not identified.
Collapse
Affiliation(s)
- Timothy F Spracklen
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Bernard Keavney
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | - Nakita Laing
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Ntobeko Ntusi
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Universities Body Imaging Centre, Cape Town, South Africa
| | - Gasnat Shaboodien
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
| |
Collapse
|
21
|
Murray B, James CA. Genotype-phenotype Correlates in Arrhythmogenic Cardiomyopathies. Curr Cardiol Rep 2022; 24:1557-1565. [PMID: 36074218 DOI: 10.1007/s11886-022-01777-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF THE REVIEW The definition of arrhythmogenic cardiomyopathy (ACM) has expanded beyond desmosomal arrhythmogenic right ventricular cardiomyopathy (ARVC) to include other genetic cardiomyopathies with a significant arrhythmia burden. Emerging data on genotype-phenotype correlations has led recent consensus guidelines to urge genetic testing as a critical component of not only diagnosis but also management of ACM. RECENT FINDINGS Plakophilin-2 (PKP2) ARVC/ACM is most likely to meet ARVC Task Force Criteria with right sided involvement and ventricular arrhythmias, while desmoplakin (DSP) ACM may have a normal electrocardiogram (ECG) and has a subepicardial LV scar pattern. Extra-desmosomal ACM including ACM associated with transmembrane protein 43 and phospholamban variants may have characteristic ECG patterns and biventricular cardiomyopathy. Lamin A/C and SCN5A cardiomyopathy often have heart block on ECG with DCM, but are distinct from DCM in that they have significantly elevated arrhythmic risk. Newer genes, especially filamin-C (FLNC) also may have distinct imaging scar patterns, arrhythmia risk, and risk predictors. Recognition of these key differences have implications for clinical management and reinforce the importance of genetic testing in the diagnosis and the emerging opportunities for genotype-specific management of ACM patients.
Collapse
Affiliation(s)
- Brittney Murray
- School of Medicine/Division of Cardiology, Johns Hopkins University, 600 N. Wolfe St. Blalock 545, Baltimore, MD, 21287, USA.
| | - Cynthia A James
- School of Medicine/Division of Cardiology, Johns Hopkins University, 600 N. Wolfe St. Blalock 545, Baltimore, MD, 21287, USA
| |
Collapse
|
22
|
Ren J, Chen L, Chen X, Zhang N, Sun X, Song J. Acylation-stimulating protein and heart failure progression in arrhythmogenic right ventricular cardiomyopathy. ESC Heart Fail 2022; 10:492-501. [PMID: 36316820 PMCID: PMC9871714 DOI: 10.1002/ehf2.14218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/14/2022] [Indexed: 01/29/2023] Open
Abstract
AIMS Our previous studies suggested that the complement system was critical in the prognosis of arrhythmogenic right ventricular cardiomyopathy (ARVC). The acylation-stimulating protein (ASP), generated through the alternate complement pathway, was reported to regulate lipogenesis and triglyceride storage. This study aimed to investigate the role of ASP in predicting adverse cardiac events in an ARVC cohort. METHODS AND RESULTS We enrolled 111 ARVC patients and 106 healthy volunteers, and measured their plasma ASP levels using enzyme-linked immunosorbent assays. Plasma ASP levels were significantly higher in the ARVC patients than in the healthy controls (2325.22 ± 20.08 vs. 2189.75 ± 15.55, P < 0.001), with a similar trend observed in the myocardial explant assay. Spearman correlation analysis indicated plasma ASP level associated with cardiac structural (right ventricular internal dimension, P = 0.006) and functional remodelling (left ventricular ejection fraction, P = 0.002) in ARVC patients. The ARVC patients were followed up for an average of 17.79 ± 1.09 months. Heart failure-associated events (HFAEs) were defined as heart transplantation, on a cardiac transplant list, or death due to end-stage heart failure. Plasma ASP levels in patients with HFAEs were significantly higher than in those without clinical events (2486.03 ± 26.70 vs. 2268.83 ± 23.51, P < 0.001) or those with malignant arrhythmic events (2486.03 ± 26.70 vs. 2297.80 ± 60.46, P = 0.008). LASSO (least absolute shrinkage and selection operator) and multivariable Cox regression analyses showed the ASP level (HR = 1.004, 95% CI [1.002,1.006], P = 0.002) was an independent predictor for adverse HFAEs in ARVC patients. The spline-fitting procedure was applied to illustrate the HFAE-free probabilities at different time points. CONCLUSIONS Our results suggest that plasma ASP may be a useful biomarker in prediction of adverse HF-associated events in ARVC patients.
Collapse
Affiliation(s)
- Jie Ren
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ningning Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaogang Sun
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
23
|
Chen Y, Sun Q, Hao C, Guo R, Wang C, Yang W, Zhang Y, Wang F, Li W, Guo J. Identification of a novel variant in N-cadherin associated with dilated cardiomyopathy. Front Med (Lausanne) 2022; 9:944950. [PMID: 36111109 PMCID: PMC9468813 DOI: 10.3389/fmed.2022.944950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Dilated cardiomyopathy (DCM), which is a major cause of heart failure, is a primary cardiac muscle disease with high morbidity and mortality rates. DCM is a genetically heritable disease and more than 10 gene ontologies have been implicated in DCM. CDH2 encodes N-cadherin and belongs to a superfamily of transmembrane proteins that mediate cell–cell adhesion in a calcium-dependent manner. Deficiency of CDH2 is associated with arrhythmogenic right ventricular cardiomyopathy (OMIM: 618920) and agenesis of the corpus callosum, cardiac, ocular, and genital syndrome (OMIM: 618929). However, there have been no reports of isolated DCM associated with CDH2 deficiency. Methods We performed whole exome sequencing in a 12-year-old girl with non-syndromic DCM and her unaffected parents. Variants in both known DCM-related genes and novel candidate genes were analyzed and pathogenicity confirmation experiments were performed. Results No pathogenic/likely pathogenic variant in known DCM-related genes was identified in the patient. We found a de novo variant in a candidate gene CDH2 in the patient, namely, c.474G>C/p.Lys158Asn (NM_001792.5). This variant has not been reported in the ClinVar or Human Gene Mutation Database (HGMD). CDH2 p.Lys158Asn was found in the conserved domain of N-cadherin, which is associated with the hydrolysis of the precursor segment and interference with adhesiveness. Furthermore, we tested the expression and efficiency of cell–cell adhesion while overexpressing the CDH2 Lys158Asn mutant and two previously reported variants in CDH2 as positive controls. The adhesion efficiency was considerably reduced in the presence of the mutated CDH2 protein compared with wild-type CDH2 protein, which suggested that the mutated CDH2 protein's adhesion capacity was impaired. The variant was probably pathogenic after integrating clinical manifestations, genetic analysis, and functional tests. Conclusion We identified a CDH2 variant in DCM. We observed a new clinical symptom associated with N-cadherin deficiency and broadened the genetic spectra of DCM.
Collapse
Affiliation(s)
- Yuanying Chen
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Qiqing Sun
- Department of Cardiology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Chanjuan Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Ruolan Guo
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Chentong Wang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Weili Yang
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Yaodong Zhang
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Fangjie Wang
- Department of Cardiology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Jun Guo
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Center of Rare Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Henan Key Laboratory of Pediatric Inherited and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
- *Correspondence: Jun Guo
| |
Collapse
|
24
|
Gao Y, Wu D, Chen B, Chen Y, Zhang Q, Zhao P. Rare Variants in Novel Candidate Genes Associated With Nonsyndromic Patent Ductus Arteriosus Identified With Whole-Exome Sequencing. Front Genet 2022; 13:921925. [PMID: 35734438 PMCID: PMC9207465 DOI: 10.3389/fgene.2022.921925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Patent ductus arteriosus (PDA) is one of the most common congenital heart defects causing pulmonary hypertension, infective endocarditis, and even death. The important role of genetics in determining spontaneous ductal closure has been well-established. However, as many of the identified variants are rare, thorough identification of the associated genetic factors is necessary to further explore the genetic etiology of PDA. Methods: We performed whole-exome sequencing (WES) on 39 isolated nonsyndromic PDA patients and 100 healthy controls. Rare variants and novel genes were identified through bioinformatic filtering strategies. The expression patterns of candidate genes were explored in human embryo heart samples. Results: Eighteen rare damaging variants of six novel PDA-associated genes (SOX8, NES, CDH2, ANK3, EIF4G1, and HIPK1) were newly identified, which were highly expressed in human embryo hearts. Conclusions: WES is an efficient diagnostic tool for exploring the genetic pathogenesis of PDA. These findings contribute new insights into the molecular basis of PDA and may inform further studies on genetic risk factors for congenital heart defects.
Collapse
Affiliation(s)
- Ying Gao
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Dan Wu
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Bo Chen
- Department of Cardiothoracic Surgery, School of Medicine, Heart Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengjun Zhao
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Pengjun Zhao,
| |
Collapse
|
25
|
Atomic Force Microscopy (AFM) Applications in Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23073700. [PMID: 35409059 PMCID: PMC8998711 DOI: 10.3390/ijms23073700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disorder characterized by progressive replacement of cardiomyocytes by fibrofatty tissue, ventricular dilatation, cardiac dysfunction, arrhythmias, and sudden cardiac death. Interest in molecular biomechanics for these disorders is constantly growing. Atomic force microscopy (AFM) is a well-established technic to study the mechanobiology of biological samples under physiological and pathological conditions at the cellular scale. However, a review which described all the different data that can be obtained using the AFM (cell elasticity, adhesion behavior, viscoelasticity, beating force, and frequency) is still missing. In this review, we will discuss several techniques that highlight the potential of AFM to be used as a tool for assessing the biomechanics involved in ACM. Indeed, analysis of genetically mutated cells with AFM reveal abnormalities of the cytoskeleton, cell membrane structures, and defects of contractility. The higher the Young’s modulus, the stiffer the cell, and it is well known that abnormal tissue stiffness is symptomatic of a range of diseases. The cell beating force and frequency provide information during the depolarization and repolarization phases, complementary to cell electrophysiology (calcium imaging, MEA, patch clamp). In addition, original data is also presented to emphasize the unique potential of AFM as a tool to assess fibrosis in cardiac tissue.
Collapse
|
26
|
Lou J, Chen H, Huang S, Chen P, Yu Y, Chen F. Update on risk factors and biomarkers of sudden unexplained cardiac death. J Forensic Leg Med 2022; 87:102332. [DOI: 10.1016/j.jflm.2022.102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023]
|
27
|
Bouska MJ, Bai H. Loxl2 is a mediator of cardiac aging in Drosophila melanogaster, genetically examining the role of aging clock genes. G3 (BETHESDA, MD.) 2022; 12:jkab381. [PMID: 34734976 PMCID: PMC8727986 DOI: 10.1093/g3journal/jkab381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/23/2021] [Indexed: 11/12/2022]
Abstract
Transcriptomic, proteomic, and methylation aging clocks demonstrate that aging has a predictable preset program, while transcriptome trajectory turning points indicate that the 20-40 age range in humans is the likely stage at which the progressive loss of homeostatic control, and in turn aging, begins to have detrimental effects. Turning points in this age range overlapping with human aging clock genes revealed five candidates that we hypothesized could play a role in aging or age-related physiological decline. To examine these gene's effects on lifespan and health-span, we utilized whole body and heart-specific gene knockdown of human orthologs in Drosophila melanogaster. Whole body lysyl oxidase like 2 (Loxl2), fz3, and Glo1 RNAi positively affected lifespan as did heart-specific Loxl2 knockdown. Loxl2 inhibition concurrently reduced age-related cardiac arrythmia and collagen (Pericardin) fiber width. Loxl2 binds several transcription factors in humans and RT-qPCR confirmed that a conserved transcriptional target CDH1 (Drosophila CadN2) has expression levels which correlate with Loxl2 reduction in Drosophila. These results point to conserved pathways and multiple mechanisms by which inhibition of Loxl2 can be beneficial to heart health and organismal aging.
Collapse
Affiliation(s)
- Mark J Bouska
- Department of Genetics, Development, & Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Hua Bai
- Department of Genetics, Development, & Cell Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
28
|
Bueno-Beti C, Asimaki A. Histopathological Features and Protein Markers of Arrhythmogenic Cardiomyopathy. Front Cardiovasc Med 2021; 8:746321. [PMID: 34950711 PMCID: PMC8688541 DOI: 10.3389/fcvm.2021.746321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable heart muscle disease characterized by syncope, palpitations, ventricular arrhythmias and sudden cardiac death (SCD) especially in young individuals. It is estimated to affect 1:5,000 individuals in the general population, with >60% of patients bearing one or more mutations in genes coding for desmosomal proteins. Desmosomes are intercellular adhesion junctions, which in cardiac myocytes reside within the intercalated disks (IDs), the areas of mechanical and electrical cell-cell coupling. Histologically, ACM is characterized by fibrofatty replacement of cardiac myocytes predominantly in the right ventricular free wall though left ventricular and biventricular forms have also been described. The disease is characterized by age-related progression, vast phenotypic manifestation and incomplete penetrance, making proband diagnosis and risk stratification of family members particularly challenging. Key protein redistribution at the IDs may represent a specific diagnostic marker but its applicability is still limited by the need for a myocardial sample. Specific markers of ACM in surrogate tissues, such as the blood and the buccal epithelium, may represent a non-invasive, safe and inexpensive alternative for diagnosis and cascade screening. In this review, we shall cover the most relevant biomarkers so far reported and discuss their potential impact on the diagnosis, prognosis and management of ACM.
Collapse
Affiliation(s)
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|
29
|
MicroRNAs: From Junk RNA to Life Regulators and Their Role in Cardiovascular Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded small non-coding RNA (18–25 nucleotides) that until a few years ago were considered junk RNA. In the last twenty years, they have acquired more importance thanks to the understanding of their influence on gene expression and their role as negative regulators at post-transcriptional level, influencing the stability of messenger RNA (mRNA). Approximately 5% of the genome encodes miRNAs which are responsible for regulating numerous signaling pathways, cellular processes and cell-to-cell communication. In the cardiovascular system, miRNAs control the functions of various cells, such as cardiomyocytes, endothelial cells, smooth muscle cells and fibroblasts, playing a role in physiological and pathological processes and seeming also related to variations in contractility and hereditary cardiomyopathies. They provide a new perspective on the pathophysiology of disorders such as hypertrophy, fibrosis, arrhythmia, inflammation and atherosclerosis. MiRNAs are differentially expressed in diseased tissue and can be released into the circulation and then detected. MiRNAs have become interesting for the development of new diagnostic and therapeutic tools for various diseases, including heart disease. In this review, the concept of miRNAs and their role in cardiomyopathies will be introduced, focusing on their potential as therapeutic and diagnostic targets (as biomarkers).
Collapse
|
30
|
Zhang K, Cloonan PE, Sundaram S, Liu F, Das SL, Ewoldt JK, Bays JL, Tomp S, Toepfer CN, Marsiglia JDC, Gorham J, Reichart D, Eyckmans J, Seidman JG, Seidman CE, Chen CS. Plakophilin-2 truncating variants impair cardiac contractility by disrupting sarcomere stability and organization. SCIENCE ADVANCES 2021; 7:eabh3995. [PMID: 34652945 PMCID: PMC8519574 DOI: 10.1126/sciadv.abh3995] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/25/2021] [Indexed: 05/10/2023]
Abstract
Progressive loss of cardiac systolic function in arrhythmogenic cardiomyopathy (ACM) has recently gained attention as an important clinical consideration in managing the disease. However, the mechanisms leading to reduction in cardiac contractility are poorly defined. Here, we use CRISPR gene editing to generate human induced pluripotent stem cells (iPSCs) that harbor plakophilin-2 truncating variants (PKP2tv), the most prevalent ACM-linked mutations. The PKP2tv iPSC–derived cardiomyocytes are shown to have aberrant action potentials and reduced systolic function in cardiac microtissues, recapitulating both the electrical and mechanical pathologies reported in ACM. By combining cell micropatterning with traction force microscopy and live imaging, we found that PKP2tvs impair cardiac tissue contractility by destabilizing cell-cell junctions and in turn disrupting sarcomere stability and organization. These findings highlight the interplay between cell-cell adhesions and sarcomeres required for stabilizing cardiomyocyte structure and function and suggest fundamental pathogenic mechanisms that may be shared among different types of cardiomyopathies.
Collapse
Affiliation(s)
- Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Paige E. Cloonan
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Subramanian Sundaram
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Feng Liu
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Shoshana L. Das
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jourdan K. Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jennifer L. Bays
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samuel Tomp
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | | | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
31
|
Rouhi L, Cheedipudi SM, Chen SN, Fan S, Lombardi R, Chen X, Coarfa C, Robertson MJ, Gurha P, Marian AJ. Haploinsufficiency of Tmem43 in cardiac myocytes activates the DNA damage response pathway leading to a late-onset senescence-associated pro-fibrotic cardiomyopathy. Cardiovasc Res 2021; 117:2377-2394. [PMID: 33070193 PMCID: PMC8861264 DOI: 10.1093/cvr/cvaa300] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/18/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
AIMS Arrhythmogenic cardiomyopathy (ACM) encompasses a genetically heterogeneous group of myocardial diseases whose manifestations are sudden cardiac death, cardiac arrhythmias, heart failure, and in a subset fibro-adipogenic infiltration of the myocardium. Mutations in the TMEM43 gene, encoding transmembrane protein 43 (TMEM43) are known to cause ACM. The purpose of the study was to gain insights into the molecular pathogenesis of ACM caused by TMEM43 haploinsufficiency. METHODS AND RESULTS The Tmem43 gene was specifically deleted in cardiac myocytes by crossing the Myh6-Cre and floxed Tmem43 mice. Myh6-Cre:Tmem43W/F mice showed an age-dependent phenotype characterized by an increased mortality, cardiac dilatation and dysfunction, myocardial fibrosis, adipogenesis, and apoptosis. Sequencing of cardiac myocyte transcripts prior to and after the onset of cardiac phenotype predicted early activation of the TP53 pathway. Increased TP53 activity was associated with increased levels of markers of DNA damage response (DDR), and a subset of senescence-associated secretary phenotype (SASP). Activation of DDR, TP53, SASP, and their selected downstream effectors, including phospho-SMAD2 and phospho-SMAD3 were validated by alternative methods, including immunoblotting. Expression of SASP was associated with epithelial-mesenchymal transition and age-dependent expression of myocardial fibrosis and apoptosis in the Myh6-Cre:Tmem43W/F mice. CONCLUSION TMEM43 haploinsufficiency is associated with activation of the DDR and the TP53 pathways, which lead to increased expression of SASP and an age-dependent expression of a pro-fibrotic cardiomyopathy. Given that TMEM43 is a nuclear envelope protein and our previous data showing deficiency of another nuclear envelope protein, namely lamin A/C, activates the DDR/TP53 pathway, we surmise that DNA damage is a shared mechanism in the pathogenesis of cardiomyopathies caused by mutations involving nuclear envelope proteins.
Collapse
Affiliation(s)
- Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Suet Nee Chen
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Raffaella Lombardi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Xiaofan Chen
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew J Robertson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| |
Collapse
|
32
|
Corrado D, Migliore F, Zorzi A. Arrhythmic risk stratification in arrhythmogenic cardiomyopathy: new predictors for left-sided variants? Eur Heart J 2021; 42:2851-2853. [PMID: 34151346 DOI: 10.1093/eurheartj/ehab355] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Domenico Corrado
- Department of Cardiac, Thoracic and Vascular Sciences, and Public Health, University of Padova, Italy
| | - Federico Migliore
- Department of Cardiac, Thoracic and Vascular Sciences, and Public Health, University of Padova, Italy
| | - Alessandro Zorzi
- Department of Cardiac, Thoracic and Vascular Sciences, and Public Health, University of Padova, Italy
| |
Collapse
|
33
|
Cheedipudi SM, Fan S, Rouhi L, Marian AJ. Pharmacological suppression of the WNT signaling pathway attenuates age-dependent expression of the phenotype in a mouse model of arrhythmogenic cardiomyopathy. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34447973 PMCID: PMC8386676 DOI: 10.20517/jca.2021.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction Arrhythmogenic cardiomyopathy (ACM) is a genetic disease of the myocardium, characterized by cardiac arrhythmias, dysfunction, and sudden cardiac death. The pathological hallmark of ACM is fibro-adipocytes replacing cardiac myocytes. The canonical WNT pathway is implicated in the pathogenesis of ACM. Aim The study aimed to determine the effects of the suppression of the WNT pathway on cardiac phenotype in a mouse model of ACM. Methods and Results One copy of the Dsp gene, a known cause of ACM in humans, was deleted specifically in cardiac myocytes (Myh6-Cre-Dsp W/F). Three-month-old wild type and Myh6-Cre-Dsp W/F mice, without a discernible phenotype, were randomized to either untreated or daily administration of a vehicle (placebo), or WNT974, the latter an established inhibitor of the WNT pathway, for three months. The Myh6-Cre-Dsp W/F mice in the untreated or placebo-treated groups exhibited cardiac dilatation and dysfunction, increased myocardial fibrosis, and apoptosis upon completion of the study, which was verified by complementary methods. Daily administration of WNT974 prevented and/or attenuated evolving cardiac dilatation and dysfunction, normalized myocardial fibrosis, and reduced apoptosis, compared to the untreated or placebo-treated groups. However, administration of WNT974 increased the number of adipocytes only in the Myh6-Cre-Dsp W/F hearts. There were no differences in the incidence of cardiac arrhythmias and survival rates. Conclusion Suppression of the WNT pathway imparts salutary phenotypic effects by preventing or attenuating age-dependent expression of cardiac dilatation and dysfunction, myocardial fibrosis, and apoptosis in a mouse model of ACM. The findings set the stage for large-scale studies and studies in larger animal models to test the beneficial effects of the suppression of the WNT pathway in ACM. One sentence summary Suppression of the WNT signaling pathway has beneficial effects on cardiac dysfunction, myocardial apoptosis, and fibrosis in a mouse model of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
34
|
Zhong A, Ding N, Zhou Y, Yang G, Peng Z, Zhang H, Chai X. Identification of Hub Genes Associated with the Pathogenesis of Intracranial Aneurysm via Integrated Bioinformatics Analysis. Int J Gen Med 2021; 14:4039-4050. [PMID: 34354366 PMCID: PMC8331219 DOI: 10.2147/ijgm.s320396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/09/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND At present, the pathogenesis of intracranial aneurysms (IA) remains unclear, which significantly hinders the development of novel strategies for the clinical treatment. In this study, bioinformatics methods were used to identify the potential hub genes and pathways associated with the pathogenesis of IA. METHODS The gene expression datasets of patients with intracranial aneurysm were downloaded from the Gene Expression Database (GEO), and the different data sets were integrated by the robust rank aggregation (RRA) method to identify the differentially expressed genes between patients with intracranial aneurysm and the controls. The functional enrichment analyses of the significant differentially expressed genes (DEGs) were performed and the protein-protein interaction (PPI) network was constructed; thereafter, the hub genes were screened by cytoHubba plug-in of Cytoscape, and finally sequencing dataset GSE122897 was used to verify the hub genes. RESULTS The GSE15629, GSE75436, GSE26969, and GSE6551 expression profiles have been included in this study, including 34 intracranial aneurysm samples and 26 control samples. The four datasets obtained 136 significant DEGs (45 up-regulated, 91 down-regulated). Enrichment analysis showed that the extracellular matrix structural constituent and the ECM-receptor interaction were closely related to the occurrence of IA. It was finally determined that eight hub genes associated with the development of IA, including VCAN, COL1A1, COL11A1, COL5A1, COL5A2, POSTN, THBS2, and CDH2. CONCLUSION The discovery of potential hub genes and pathways could enhance the understanding of the molecular mechanisms associated with the development of IA. These hub genes may be potential therapeutic targets for the management and new biomarker for the diagnosis of IA.
Collapse
Affiliation(s)
- Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Ning Ding
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yang Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhenyu Peng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hongliang Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Trauma center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
35
|
Migliore F, Mattesi G, Zorzi A, Bauce B, Rigato I, Corrado D, Cipriani A. Arrhythmogenic Cardiomyopathy-Current Treatment and Future Options. J Clin Med 2021; 10:2750. [PMID: 34206637 PMCID: PMC8268983 DOI: 10.3390/jcm10132750] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inheritable heart muscle disease characterised pathologically by fibrofatty myocardial replacement and clinically by ventricular arrhythmias (VAs) and sudden cardiac death (SCD). Although, in its original description, the disease was believed to predominantly involve the right ventricle, biventricular and left-dominant variants, in which the myocardial lesions affect in parallel or even mostly the left ventricle, are nowadays commonly observed. The clinical management of these patients has two main purposes: the prevention of SCD and the control of arrhythmic and heart failure (HF) events. An implantable cardioverter defibrillator (ICD) is the only proven lifesaving treatment, despite significant morbidity because of device-related complications and inappropriate shocks. Selection of patients who can benefit the most from ICD therapy is one of the most challenging issues in clinical practice. Risk stratification in ACM patients is mostly based on arrhythmic burden and ventricular dysfunction severity, although other clinical features resulting from electrocardiogram and imaging modalities such as cardiac magnetic resonance may have a role. Medical therapy is crucial for treatment of VAs and the prevention of negative ventricular remodelling. In this regard, the efficacy of novel anti-HF molecules and drugs acting on the inflammatory pathway in patients with ACM is, to date, unknown. Catheter ablation represents an effective strategy to treat ventricular tachycardia relapses and recurrent ICD shocks. The present review will address the current strategies for prevention of SCD and treatment of VAs and HF in patients with ACM.
Collapse
Affiliation(s)
- Federico Migliore
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Giulia Mattesi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alessandro Zorzi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Barbara Bauce
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Ilaria Rigato
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Domenico Corrado
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alberto Cipriani
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
36
|
Kraus SM, Shaboodien G, Francis V, Laing N, Cirota J, Chin A, Pandie S, Lawrenson J, Comitis GAM, Fourie B, Zühlke L, Wonkam A, Wainwright H, Damasceno A, Mocumbi AO, Pepeta L, Moeketsi K, Thomas BM, Thomas K, Makotoko M, Brown S, Ntsekhe M, Sliwa K, Badri M, Gumedze F, Cordell HJ, Keavney B, Ferreira V, Mahmod M, Cooper LT, Yacoub M, Neubauer S, Watkins H, Mayosi BM, Ntusi NAB. Rationale and design of the African Cardiomyopathy and Myocarditis Registry Program: The IMHOTEP study. Int J Cardiol 2021; 333:119-126. [PMID: 33607192 DOI: 10.1016/j.ijcard.2021.02.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Heart failure (HF), the dominant form of cardiovascular disease in Africans, is mainly due to hypertension, rheumatic heart disease and cardiomyopathy. Cardiomyopathies pose a great challenge because of poor prognosis and high prevalence in low- and middle-income countries (LMICs). Little is known about the etiology and outcome of cardiomyopathy in Africa. Specifically, the role of myocarditis and the genetic causes of cardiomyopathy are largely unidentified in Africans. METHOD The African Cardiomyopathy and Myocarditis Registry Program (the IMHOTEP study) is a pan-African multi-centre, hospital-based cohort study, designed with the primary aim of describing the clinical characteristics, genetic causes, prevalence, management and outcome of cardiomyopathy and myocarditis in children and adults. The secondary aim is to identify barriers to the implementation of evidence-based care and provide a platform for trials and other intervention studies to reduce morbidity and mortality in cardiomyopathy. The registry consists of a prospective cohort of newly diagnosed (i.e., incident) cases and a retrospective (i.e., prevalent) cohort of existing cases from participating centres. Patients with cardiomyopathy and myocarditis will be subjected to a standardized 3-stage diagnostic process. To date, 750 patients have been recruited into the multi-centre pilot phase of the study. CONCLUSION The IMHOTEP study will provide comprehensive and novel data on clinical features, genetic causes, prevalence and outcome of African children and adults with all forms of cardiomyopathy and myocarditis in Africa. Based on these findings, appropriate strategies for management and prevention of the cardiomyopathies in LMICs are likely to emerge.
Collapse
Affiliation(s)
- Sarah M Kraus
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Gasnat Shaboodien
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Veronica Francis
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Nakita Laing
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa; Division of Human Genetics, Department of Medicine, UCT, Cape Town, South Africa
| | - Jacqui Cirota
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Ashley Chin
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Shahiemah Pandie
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - John Lawrenson
- Division of Paediatric Cardiology, Department of Paediatrics and Child Health, UCT and Red Cross War Memorial Children's Hospital, Cape Town, South Africa; Division of Paediatric Cardiology, Department of Paediatrics and Child Health, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - George A M Comitis
- Division of Paediatric Cardiology, Department of Paediatrics and Child Health, UCT and Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| | - Barend Fourie
- Division of Paediatric Cardiology, Department of Paediatrics and Child Health, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Liesl Zühlke
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa; Division of Paediatric Cardiology, Department of Paediatrics and Child Health, UCT and Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| | - Ambroise Wonkam
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa; Division of Human Genetics, Department of Medicine, UCT, Cape Town, South Africa
| | - Helen Wainwright
- Department of Pathology, National Health Laboratory Service and UCT, Cape Town, South Africa
| | | | - Ana Olga Mocumbi
- Instituto Nacional de Saúde and Eduardo Mondlane University, Maputo, Mozambique
| | - Lungile Pepeta
- Department of Paediatrics, Port Elizabeth Hospital Complex and Nelson Mandela Metropolitan University, Port Elizabeth, South Africa
| | - Khulile Moeketsi
- Division of Cardiology, Nelson Mandela Academic Hospital and Walter Sisulu University, Mthatha, South Africa
| | - Baby M Thomas
- Division of Cardiology, Nelson Mandela Academic Hospital and Walter Sisulu University, Mthatha, South Africa
| | - Kandathil Thomas
- Division of Cardiology, Nelson Mandela Academic Hospital and Walter Sisulu University, Mthatha, South Africa
| | - Makoali Makotoko
- Division of Cardiology, Universitas Hospital and University of the Free State, Bloemfontein, South Africa
| | - Stephen Brown
- Division of Cardiology, Universitas Hospital and University of the Free State, Bloemfontein, South Africa
| | - Mpiko Ntsekhe
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Karen Sliwa
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Motasim Badri
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa; College of Medicine, King Saudi Bin Abdulaziz University for Medical Sciences, Riyadh, Saudi Arabia
| | | | - Heather J Cordell
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bernard Keavney
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Vanessa Ferreira
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Masliza Mahmod
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, USA
| | | | - Stefan Neubauer
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bongani M Mayosi
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa
| | - Ntobeko A B Ntusi
- The Cardiac Clinic and Hatter Institute of Cardiovascular Research in Africa, Department of Medicine, University of Cape Town (UCT) and Groote Schuur Hospital, Cape Town, South Africa.
| |
Collapse
|
37
|
Variants in MHY7 Gene Cause Arrhythmogenic Cardiomyopathy. Genes (Basel) 2021; 12:genes12060793. [PMID: 34067482 PMCID: PMC8224781 DOI: 10.3390/genes12060793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Arrhythmogenic Cardiomyopathy (ACM) is a disease of the cardiac muscle, characterized by frequent ventricular arrhythmias and functional/ structural abnormalities, mainly of the right ventricle. To date, 20 different genes have been associated with ACM and the majority of them encode for desmosomal proteins. In this study, we describe the characterization of two novel variants in MHY7 gene, segregating in two ACM families. MYH7 encodes for myosin heavy chain β (MHC-β) isoform, involved in cardiac muscle contractility. METHOD AND RESULTS In family A, the autopsy revealed ACM with biventricular involvement in both the proband and his father. In family B, the proband had been diagnosed as affected by ACM and implanted with implantable cardioverter defibrillator (ICD), due to ECG evidence of monomorphic ventricular tachycardia after syncope. After clinical evaluation, a molecular diagnosis was performed using a NGS custom panel. The two novel variants identified predicted damaging, located in a highly conserved domain: c. 2630T>C is not described while c.2609G>A has a frequency of 0.00000398. In silico analyses evaluated the docking characteristics between proteins using the Haddock2.2 webserver. CONCLUSIONS Our results reveal two variants in sarcomeric genes to be the molecular cause of ACM, further increasing the genetic heterogeneity of the disease; in fact, sarcomeric variants are usually associated with HCM phenotype. Studies on the role of sarcomere genes in the pathogenesis of ACM are surely recommended in those ACM patients negative for desmosomal mutation screening.
Collapse
|
38
|
Ben-Haim Y, Asimaki A, Behr ER. Brugada syndrome and arrhythmogenic cardiomyopathy: overlapping disorders of the connexome? Europace 2021; 23:653-664. [PMID: 33200179 DOI: 10.1093/europace/euaa277] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/19/2020] [Indexed: 12/19/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) and Brugada syndrome (BrS) are inherited diseases characterized by an increased risk for arrhythmias and sudden cardiac death. Possible overlap between the two was suggested soon after the description of BrS. Since then, various studies focusing on different aspects have been published pointing to similar findings in the two diseases. More recent findings on the structure of the cardiac cell-cell junctions may unite the pathophysiology of both diseases and give further evidence to the theory that they may in part be variants of the same disease spectrum. In this review, we aim to summarize the studies indicating the pathophysiological, genetic, structural, and electrophysiological overlap between ACM and BrS.
Collapse
Affiliation(s)
- Yael Ben-Haim
- Institute of Molecular and Clinical Sciences, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
- Cardiology Clinical Academic Group, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - Angeliki Asimaki
- Institute of Molecular and Clinical Sciences, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
- Cardiology Clinical Academic Group, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - Elijah R Behr
- Institute of Molecular and Clinical Sciences, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
- Cardiology Clinical Academic Group, St. George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
39
|
Mattesi G, Cipriani A, Bauce B, Rigato I, Zorzi A, Corrado D. Arrhythmogenic Left Ventricular Cardiomyopathy: Genotype-Phenotype Correlations and New Diagnostic Criteria. J Clin Med 2021; 10:jcm10102212. [PMID: 34065276 PMCID: PMC8160676 DOI: 10.3390/jcm10102212] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease characterized by loss of ventricular myocardium and fibrofatty replacement, which predisposes to scar-related ventricular arrhythmias and sudden cardiac death, particularly in the young and athletes. Although in its original description the disease was characterized by an exclusive or at least predominant right ventricle (RV) involvement, it has been demonstrated that the fibrofatty scar can also localize in the left ventricle (LV), with the LV lesion that can equalize or even overcome that of the RV. While the right-dominant form is typically associated with mutations in genes encoding for desmosomal proteins, other (non-desmosomal) mutations have been showed to cause the biventricular and left-dominant variants. This has led to a critical evaluation of the 2010 International Task Force criteria, which exclusively addressed the right phenotypic manifestations of ACM. An International Expert consensus document has been recently developed to provide upgraded criteria (“the Padua Criteria”) for the diagnosis of the whole spectrum of ACM phenotypes, particularly left-dominant forms, highlighting the use of cardiac magnetic resonance. This review aims to offer an overview of the current knowledge on the genetic basis, the phenotypic expressions, and the diagnosis of left-sided variants, both biventricular and left-dominant, of ACM.
Collapse
|
40
|
Qiu X, Lin J, Liang B, Chen Y, Liu G, Zheng J. Identification of Hub Genes and MicroRNAs Associated With Idiopathic Pulmonary Arterial Hypertension by Integrated Bioinformatics Analyses. Front Genet 2021; 12:667406. [PMID: 33995494 PMCID: PMC8117102 DOI: 10.3389/fgene.2021.636934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
Objective The aim of this study is the identification of hub genes associated with idiopathic pulmonary arterial hypertension (IPAH). Materials and Methods GSE15197 gene expression data was downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified by screening IPAH patients and controls. The 5,000 genes with the greatest variances were analyzed using a weighted gene co-expression network analysis (WGCNA). Modules with the strongest correlation with IPAH were chosen, followed by a functional enrichment analysis. Protein–protein interaction (PPI) networks were constructed to identify hub gene candidates using calculated degrees. Real hub genes were found from the overlap of DEGs and candidate hub genes. microRNAs (miRNAs) targeting real hub genes were found by screening miRNet 2.0. The most important IPAH miRNAs were identified. Results There were 4,395 DEGs identified. WGCNA indicated that green and brown modules associated most strongly with IPAH. Functional enrichment analysis showed that green and brown module genes were mainly involved in protein digestion and absorption and proteoglycans in cancer, respectively. The top ten candidate hub genes in green and brown modules were identified, respectively. After overlapping with DEGs, 11 real hub genes were identified: EP300, MMP2, CDH2, CDK2, GNG10, ALB, SMC2, DHX15, CUL3, BTBD1, and LTN1. These genes were expressed with significant differences in IPAH versus controls, indicating a high diagnostic ability. The miRNA–gene network showed that hsa-mir-1-3p could associate with IPAH. Conclusion EP300, MMP2, CDH2, CDK2, GNG10, ALB, SMC2, DHX15, CUL3, BTBD1, and LTN1 may play essential roles in IPAH. Predicted miRNA hsa-mir-1-3p could regulate gene expression in IPAH. Such hub genes may contribute to the pathology and progression in IPAH, providing potential diagnostic and therapeutic opportunities for IPAH patients.
Collapse
Affiliation(s)
- Xue Qiu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinyan Lin
- The First Clinical Medical School, Guangxi Medical University, Nanning, China
| | - Bixiao Liang
- The First Clinical Medical School, Guangxi Medical University, Nanning, China
| | - Yanbing Chen
- The First Clinical Medical School, Guangxi Medical University, Nanning, China
| | - Guoqun Liu
- The First Clinical Medical School, Guangxi Medical University, Nanning, China
| | - Jing Zheng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
41
|
Dworschak GC, Reutter HM, Ludwig M. Currarino syndrome: a comprehensive genetic review of a rare congenital disorder. Orphanet J Rare Dis 2021; 16:167. [PMID: 33836786 PMCID: PMC8034116 DOI: 10.1186/s13023-021-01799-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/30/2021] [Indexed: 11/10/2022] Open
Abstract
Background The triad of a presacral mass, sacral agenesis and an anorectal anomaly constitutes the rare Currarino syndrome (CS), which is caused by dorsal–ventral patterning defects during embryonic development. The major causative CS gene is MNX1, encoding a homeobox protein. Main body In the majority of patients, CS occurs as an autosomal dominant trait; however, a female predominance observed, implies that CS may underlie an additional mode(s) of inheritance. Often, the diagnosis of CS is established solely by clinical findings, impacting a detailed analysis of the disease. Our combined data, evaluating more than 60 studies reporting patients with CS-associated mutations, revealed a slightly higher incidence rate in females with a female-to-male ratio of 1.39:1. Overall, MNX1 mutation analysis was successful in only 57.4% of all CS patients investigated, with no mutation detected in 7.7% of the familial and 68% of the sporadic patients. Our studies failed to detect the presence of an expressed MNX1 isoform that might explain at least some of these mutation-negative cases. Conclusion Aside from MNX1, other genes or regulatory regions may contribute to CS and we discuss several cytogenetic studies and whole-exome sequencing data that have implicated further loci/genes in its etiology. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01799-0.
Collapse
Affiliation(s)
- Gabriel C Dworschak
- Institute of Human Genetics, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany. .,Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, 53115, Bonn, Germany. .,Department of Pediatrics, University Hospital Bonn, 53127, Bonn, Germany.
| | - Heiko M Reutter
- Institute of Human Genetics, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.,Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, 53127, Bonn, Germany
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53127, Bonn, Germany
| |
Collapse
|
42
|
James CA, Jongbloed JDH, Hershberger RE, Morales A, Judge DP, Syrris P, Pilichou K, Domingo AM, Murray B, Cadrin-Tourigny J, Lekanne Deprez R, Celeghin R, Protonotarios A, Asatryan B, Brown E, Jordan E, McGlaughon J, Thaxton C, Kurtz CL, van Tintelen JP. International Evidence Based Reappraisal of Genes Associated With Arrhythmogenic Right Ventricular Cardiomyopathy Using the Clinical Genome Resource Framework. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003273. [PMID: 33831308 PMCID: PMC8205996 DOI: 10.1161/circgen.120.003273] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited disease characterized by ventricular arrhythmias and progressive ventricular dysfunction. Genetic testing is recommended, and a pathogenic variant in an ARVC-associated gene is a major criterion for diagnosis according to the 2010 Task Force Criteria. As incorrect attribution of a gene to ARVC can contribute to misdiagnosis, we assembled an international multidisciplinary ARVC Clinical Genome Resource Gene Curation Expert Panel to reappraise all reported ARVC genes. METHODS Following a comprehensive literature search, six 2-member teams conducted blinded independent curation of reported ARVC genes using the semiquantitative Clinical Genome Resource framework. RESULTS Of 26 reported ARVC genes, only 6 (PKP2, DSP, DSG2, DSC2, JUP, and TMEM43) had strong evidence and were classified as definitive for ARVC causation. There was moderate evidence for 2 genes, DES and PLN. The remaining 18 genes had limited or no evidence. RYR2 was refuted as an ARVC gene since clinical data and model systems exhibited a catecholaminergic polymorphic ventricular tachycardia phenotype. In ClinVar, only 5 pathogenic/likely pathogenic variants (1.1%) in limited evidence genes had been reported in ARVC cases in contrast to 450 desmosome gene variants (97.4%). CONCLUSIONS Using the Clinical Genome Resource approach to gene-disease curation, only 8 genes (PKP2, DSP, DSG2, DSC2, JUP, TMEM43, PLN, and DES) had definitive or moderate evidence for ARVC, and these genes accounted for nearly all pathogenic/likely pathogenic ARVC variants in ClinVar. Therefore, only pathogenic/likely pathogenic variants in these 8 genes should yield a major criterion for ARVC diagnosis. Pathogenic/likely pathogenic variants identified in other genes in a patient should prompt further phenotyping as variants in many of these genes are associated with other cardiovascular conditions.
Collapse
Affiliation(s)
- Cynthia A James
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD (C.A.J., B.M., E.B.)
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, the Netherlands (J.D.H.J.)
| | - Ray E Hershberger
- Division of Cardiovascular Medicine, Department of Internal Medicine (R.E.H., E.J.), Ohio State University, Columbus.,Division of Human Genetics, Department of Internal Medicine (R.E.H., A.M.), Ohio State University, Columbus
| | - Ana Morales
- Division of Human Genetics, Department of Internal Medicine (R.E.H., A.M.), Ohio State University, Columbus
| | - Daniel P Judge
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston (D.P.J.)
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (P.S., A.P.)
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padua, Italy (K.P., R.C.)
| | - Argelia Medeiros Domingo
- Department for Cardiology, Inselspital, Bern University Hospital, University of Bern, Switzerland (A.M.D., B.A.)
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD (C.A.J., B.M., E.B.)
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Centre, Montreal Heart Institute, Université de Montréal, Canada (J.C.-T.)
| | - Ronald Lekanne Deprez
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, the Netherlands (R.L.D., J.P.v.T.)
| | - Rudy Celeghin
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padua, Italy (K.P., R.C.)
| | - Alexandros Protonotarios
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (P.S., A.P.)
| | - Babken Asatryan
- Department for Cardiology, Inselspital, Bern University Hospital, University of Bern, Switzerland (A.M.D., B.A.)
| | - Emily Brown
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD (C.A.J., B.M., E.B.)
| | - Elizabeth Jordan
- Division of Cardiovascular Medicine, Department of Internal Medicine (R.E.H., E.J.), Ohio State University, Columbus
| | - Jennifer McGlaughon
- Department of Genetics, University of North Carolina, Chapel Hill (J.M., C.T., C.L.K.)
| | - Courtney Thaxton
- Department of Genetics, University of North Carolina, Chapel Hill (J.M., C.T., C.L.K.)
| | - C Lisa Kurtz
- Department of Genetics, University of North Carolina, Chapel Hill (J.M., C.T., C.L.K.)
| | - J Peter van Tintelen
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, the Netherlands (R.L.D., J.P.v.T.).,Department of Genetics, University of Utrecht, University Medical Center Utrecht, the Netherlands (J.P.v.T.)
| |
Collapse
|
43
|
Volani C, Rainer J, Hernandes VV, Meraviglia V, Pramstaller PP, Smárason SV, Pompilio G, Casella M, Sommariva E, Paglia G, Rossini A. Metabolic Signature of Arrhythmogenic Cardiomyopathy. Metabolites 2021; 11:metabo11040195. [PMID: 33805952 PMCID: PMC8064316 DOI: 10.3390/metabo11040195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/06/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic-based cardiac disease accompanied by severe ventricular arrhythmias and a progressive substitution of the myocardium with fibro-fatty tissue. ACM is often associated with sudden cardiac death. Due to the reduced penetrance and variable expressivity, the presence of a genetic defect is not conclusive, thus complicating the diagnosis of ACM. Recent studies on human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) obtained from ACM individuals showed a dysregulated metabolic status, leading to the hypothesis that ACM pathology is characterized by an impairment in the energy metabolism. However, despite efforts having been made for the identification of ACM specific biomarkers, there is still a substantial lack of information regarding the whole metabolomic profile of ACM patients. The aim of the present study was to investigate the metabolic profiles of ACM patients compared to healthy controls (CTRLs). The targeted Biocrates AbsoluteIDQ® p180 assay was used on plasma samples. Our analysis showed that ACM patients have a different metabolome compared to CTRLs, and that the pathways mainly affected include tryptophan metabolism, arginine and proline metabolism and beta oxidation of fatty acids. Altogether, our data indicated that the plasma metabolomes of arrhythmogenic cardiomyopathy patients show signs of endothelium damage and impaired nitric oxide (NO), fat, and energy metabolism.
Collapse
Affiliation(s)
- Chiara Volani
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
- Correspondence:
| | - Johannes Rainer
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Vinicius Veri Hernandes
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Viviana Meraviglia
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Peter Paul Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Sigurður Vidir Smárason
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milan, Italy; (G.P.); (E.S.)
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20138 Milan, Italy
| | - Michela Casella
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
- Cardiology and Arrhythmology Clinic, University Hospital Ospedali Riuniti Umberto I-Lancisi-Salesi, 60126 Ancona, Italy
- Department of Clinical, Special and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milan, Italy; (G.P.); (E.S.)
| | - Giuseppe Paglia
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, 20854 Vedano al Lambro, Italy;
| | - Alessandra Rossini
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| |
Collapse
|
44
|
Ghidoni A, Elliott PM, Syrris P, Calkins H, James CA, Judge DP, Murray B, Barc J, Probst V, Schott JJ, Song JP, Hauer RNW, Hoorntje ET, van Tintelen JP, Schulze-Bahr E, Hamilton RM, Mittal K, Semsarian C, Behr ER, Ackerman MJ, Basso C, Parati G, Gentilini D, Kotta MC, Mayosi BM, Schwartz PJ, Crotti L. Cadherin 2-Related Arrhythmogenic Cardiomyopathy: Prevalence and Clinical Features. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003097. [PMID: 33566628 DOI: 10.1161/circgen.120.003097] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac disease characterized by fibrofatty replacement of the right and left ventricle, often causing ventricular dysfunction and life-threatening arrhythmias. Variants in desmosomal genes account for up to 60% of cases. Our objective was to establish the prevalence and clinical features of ACM stemming from pathogenic variants in the nondesmosomal cadherin 2 (CDH2), a novel genetic substrate of ACM. METHODS A cohort of 500 unrelated patients with a definite diagnosis of ACM and no disease-causing variants in the main ACM genes was assembled. Genetic screening of CDH2 was performed through next-generation or Sanger sequencing. Whenever possible, cascade screening was initiated in the families of CDH2-positive probands, and clinical evaluation was performed. RESULTS Genetic screening of CDH2 led to the identification of 7 rare variants: 5, identified in 6 probands, were classified as pathogenic or likely pathogenic. The previously established p.D407N pathogenic variant was detected in 2 additional probands. Probands and family members with pathogenic/likely pathogenic variants in CDH2 were clinically evaluated, and along with previously published cases, altogether contributed to the identification of gene-specific features (13 cases from this cohort and 11 previously published, for a total of 9 probands and 15 family members). Ventricular arrhythmic events occurred in most CDH2-positive subjects (20/24, 83%), while the occurrence of heart failure was rare (2/24, 8.3%). Among probands, sustained ventricular tachycardia and sudden cardiac death occurred in 5/9 (56%). CONCLUSIONS In this worldwide cohort of previously genotype-negative ACM patients, the prevalence of probands with CDH2 pathogenic/likely pathogenic variants was 1.2% (6/500). Our data show that this cohort of CDH2-ACM patients has a high incidence of ventricular arrhythmias, while evolution toward heart failure is rare.
Collapse
Affiliation(s)
- Alice Ghidoni
- Center for Cardiac Arrhythmias of Genetic Origin (A.G., M.-C.K., P.J.S., L.C.), Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Perry M Elliott
- Center for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (P.M.E., P.S.)
| | - Petros Syrris
- Center for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (P.M.E., P.S.)
| | - Hugh Calkins
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (H.C., C.A.J., B.M.)
| | - Cynthia A James
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (H.C., C.A.J., B.M.)
| | - Daniel P Judge
- Medical University of South Carolina, Charleston, SC (D.P.J.)
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (H.C., C.A.J., B.M.)
| | - Julien Barc
- Université de Nantes (J.B.), CNRS, Inserm, l'Institut du Thorax, France
| | - Vincent Probst
- Université de Nantes, CHU Nantes (V.P., J.J.S.), CNRS, Inserm, l'Institut du Thorax, France.,Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.)
| | - Jean Jacques Schott
- Université de Nantes, CHU Nantes (V.P., J.J.S.), CNRS, Inserm, l'Institut du Thorax, France
| | - Jiang-Ping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.-P.S.)
| | - Richard N W Hauer
- Netherlands Heart Institute (R.N.W.H., E.T.H., J.P.v.T.), University Medical Center Utrecht.,Department of Cardiology (R.N.W.H.), University Medical Center Utrecht
| | - Edgar T Hoorntje
- Netherlands Heart Institute (R.N.W.H., E.T.H., J.P.v.T.), University Medical Center Utrecht.,Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands (E.T.H.)
| | - J Peter van Tintelen
- Netherlands Heart Institute (R.N.W.H., E.T.H., J.P.v.T.), University Medical Center Utrecht.,Department of Genetics (J.P.v.T.), University Medical Center Utrecht
| | - Eric Schulze-Bahr
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.).,Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (E.S.-B.)
| | | | - Kirti Mittal
- Hospital for Sick Children, Toronto, ON, Canada (R.M.H., K.M.)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, NSW, Australia (C.S.)
| | - Elijah R Behr
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.).,Cardiology Clinical Academic Group, Institute of Molecular and Clinical Sciences, St George's University of London, St George's University Hospitals NHS Foundation Trust, London, United Kingdom (E.R.B.)
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine (Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic), Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), and Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN (M.J.A.)
| | - Cristina Basso
- Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.).,Cardiovascular Pathology Unit, Department of Cardiac, Thoracic and Vascular Sciences, University and Hospital of Padua, Italy (C.B.)
| | - Gianfranco Parati
- Istituto Auxologico Italiano, IRCCS, Cardiomyopathies Unit, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Milan (G.P., L.C.).,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., L.C.)
| | - Davide Gentilini
- Bioinformatics and Statistical Genomics Unit (D.G.), Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Italy (D.G.)
| | - Maria-Christina Kotta
- Center for Cardiac Arrhythmias of Genetic Origin (A.G., M.-C.K., P.J.S., L.C.), Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Bongani M Mayosi
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa, Groote Schuur Hospital and Division of Cardiology, Faculty of Health Sciences, University of Cape Town, South Africa (B.M.M.)
| | - Peter J Schwartz
- Center for Cardiac Arrhythmias of Genetic Origin (A.G., M.-C.K., P.J.S., L.C.), Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.)
| | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin (A.G., M.-C.K., P.J.S., L.C.), Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Member of the European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart (V.P., E.S.-B., E.R.B., C.B., P.J.S., L.C.).,Istituto Auxologico Italiano, IRCCS, Cardiomyopathies Unit, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Milan (G.P., L.C.).,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., L.C.)
| |
Collapse
|
45
|
Towbin JA, McKenna WJ, Abrams DJ, Ackerman MJ, Calkins H, Darrieux FCC, Daubert JP, de Chillou C, DePasquale EC, Desai MY, Estes NAM, Hua W, Indik JH, Ingles J, James CA, John RM, Judge DP, Keegan R, Krahn AD, Link MS, Marcus FI, McLeod CJ, Mestroni L, Priori SG, Saffitz JE, Sanatani S, Shimizu W, van Tintelen JP, Wilde AAM, Zareba W. 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy: Executive summary. Heart Rhythm 2020; 16:e373-e407. [PMID: 31676023 DOI: 10.1016/j.hrthm.2019.09.019] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 01/14/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an arrhythmogenic disorder of the myocardium not secondary to ischemic, hypertensive, or valvular heart disease. ACM incorporates a broad spectrum of genetic, systemic, infectious, and inflammatory disorders. This designation includes, but is not limited to, arrhythmogenic right/left ventricular cardiomyopathy, cardiac amyloidosis, sarcoidosis, Chagas disease, and left ventricular noncompaction. The ACM phenotype overlaps with other cardiomyopathies, particularly dilated cardiomyopathy with arrhythmia presentation that may be associated with ventricular dilatation and/or impaired systolic function. This expert consensus statement provides the clinician with guidance on evaluation and management of ACM and includes clinically relevant information on genetics and disease mechanisms. PICO questions were utilized to evaluate contemporary evidence and provide clinical guidance related to exercise in arrhythmogenic right ventricular cardiomyopathy. Recommendations were developed and approved by an expert writing group, after a systematic literature search with evidence tables, and discussion of their own clinical experience, to present the current knowledge in the field. Each recommendation is presented using the Class of Recommendation and Level of Evidence system formulated by the American College of Cardiology and the American Heart Association and is accompanied by references and explanatory text to provide essential context. The ongoing recognition of the genetic basis of ACM provides the opportunity to examine the diverse triggers and potential common pathway for the development of disease and arrhythmia.
Collapse
Affiliation(s)
- Jeffrey A Towbin
- Le Bonheur Children's Hospital, Memphis, Tennessee; University of Tennessee Health Science Center, Memphis, Tennessee
| | - William J McKenna
- University College London, Institute of Cardiovascular Science, London, United Kingdom
| | | | | | | | | | | | | | | | | | - N A Mark Estes
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Wei Hua
- Fu Wai Hospital, Beijing, China
| | - Julia H Indik
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | - Jodie Ingles
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
| | | | - Roy M John
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel P Judge
- Medical University of South Carolina, Charleston, South Carolina
| | - Roberto Keegan
- Hospital Privado Del Sur, Buenos Aires, Argentina; Hospital Español, Bahia Blanca, Argentina
| | | | - Mark S Link
- UT Southwestern Medical Center, Dallas, Texas
| | - Frank I Marcus
- University of Arizona, Sarver Heart Center, Tucson, Arizona
| | | | - Luisa Mestroni
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Silvia G Priori
- University of Pavia, Pavia, Italy; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); ICS Maugeri, IRCCS, Pavia, Italy
| | | | | | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - J Peter van Tintelen
- University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Utrecht University Medical Center Utrecht, University of Utrecht, Department of Genetics, Utrecht, the Netherlands
| | - Arthur A M Wilde
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart); University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands; Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | | |
Collapse
|
46
|
Beffagna G, Sommariva E, Bellin M. Mechanotransduction and Adrenergic Stimulation in Arrhythmogenic Cardiomyopathy: An Overview of in vitro and in vivo Models. Front Physiol 2020; 11:568535. [PMID: 33281612 PMCID: PMC7689294 DOI: 10.3389/fphys.2020.568535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/19/2020] [Indexed: 01/09/2023] Open
Abstract
Arrhythmogenic Cardiomyopathy (AC) is a rare inherited heart disease, manifesting with progressive myocardium degeneration and dysfunction, and life-threatening arrhythmic events that lead to sudden cardiac death. Despite genetic determinants, most of AC patients admitted to hospital are athletes or very physically active people, implying the existence of other disease-causing factors. It is recognized that AC phenotypes are enhanced and triggered by strenuous physical activity, while excessive mechanical stretch and load, and repetitive adrenergic stimulation are mechanisms influencing disease penetrance. Different approaches have been undertaken to recapitulate and study both mechanotransduction and adrenergic signaling in AC, including the use of in vitro cellular and tissue models, and the development of in vivo models (particularly rodents but more recently also zebrafish). However, it remains challenging to reproduce mechanical load stimuli and physical activity in laboratory experimental settings. Thus, more work to drive the innovation of advanced AC models is needed to recapitulate these subtle physiological influences. Here, we review the state-of-the-art in this field both in clinical and laboratory-based modeling scenarios. Specific attention will be focused on highlighting gaps in the knowledge and how they may be resolved by utilizing novel research methodology.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.,Department of Biology, University of Padua, Padua, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Milena Bellin
- Department of Biology, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
47
|
Sun T, Huang GY, Wang ZH, Teng SH, Cao YH, Sun JL, Hanif Q, Chen NB, Lei CZ, Liao YY. Selection signatures of Fuzhong Buffalo based on whole-genome sequences. BMC Genomics 2020; 21:674. [PMID: 32993537 PMCID: PMC7526191 DOI: 10.1186/s12864-020-07095-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fuzhong buffalo, a native breed of Guangxi Zhuang Autonomous Region, is traditionally used as a draft animal to provide farm power in the rice cultivation. In addition, the Fuzhong buffalo also prepared for the bullfighting festival organized by the locals. The detection of the selective signatures in its genome can help in elucidating the selection mechanisms in its stamina and muscle development of a draft animal. RESULTS In this study, we analyzed 27 whole genomes of buffalo (including 15 Fuzhong buffalo genomes and 12 published buffalo genomes from Upper Yangtze region). The ZHp, ZFst, π-Ratio, and XP-EHH statistics were used to identify the candidate signatures of positive selection in Fuzhong buffalo. Our results detected a set of candidate genes involving in the pathways and GO terms associated with the response to exercise (e.g., ALDOA, STAT3, AKT2, EIF4E2, CACNA2D2, TCF4, CDH2), immunity (e.g., PTPN22, NKX2-3, PIK3R1, ITK, TMEM173), nervous system (e.g., PTPN21, ROBO1, HOMER1, MAGI2, SLC1A3, NRG3, SNAP47, CTNNA2, ADGRL3). In addition, we also identified several genes related to production and growth traits (e.g., PHLPP1, PRKN, MACF1, UCN3, RALGAPA1, PHKB, PKD1L). Our results depicted several pathways, GO terms, and candidate genes to be associated with response to exercise, immunity, nervous system, and growth traits. CONCLUSIONS The selective sweep analysis of the Fuzhong buffalo demonstrated positive selection pressure on potential target genes involved in behavior, immunity, and growth traits, etc. Our findings provided a valuable resource for future research on buffalo breeding and an insight into the mechanisms of artificial selection.
Collapse
Affiliation(s)
- Ting Sun
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Guang-Yun Huang
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China
| | - Zi-Hao Wang
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China
| | - Shao-Hua Teng
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China
| | - Yan-Hong Cao
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China
| | - Jun-Li Sun
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China
| | - Quratulain Hanif
- Computational Biology Laboratory, Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan.,Department of Biotechnology, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad, Pakistan
| | - Ning-Bo Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chu-Zhao Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Yu-Ying Liao
- Animal Husbandry Institute of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Livestock Genetic Improvement, Nanning, 530001, China.
| |
Collapse
|
48
|
Patel V, Asatryan B, Siripanthong B, Munroe PB, Tiku-Owens A, Lopes LR, Khanji MY, Protonotarios A, Santangeli P, Muser D, Marchlinski FE, Brady PA, Chahal CAA. State of the Art Review on Genetics and Precision Medicine in Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21186615. [PMID: 32927679 PMCID: PMC7554944 DOI: 10.3390/ijms21186615] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterised by ventricular arrhythmia and an increased risk of sudden cardiac death (SCD). Numerous genetic determinants and phenotypic manifestations have been discovered in ACM, posing a significant clinical challenge. Further to this, wider evaluation of family members has revealed incomplete penetrance and variable expressivity in ACM, suggesting a complex genotype-phenotype relationship. This review details the genetic basis of ACM with specific genotype-phenotype associations, providing the reader with a nuanced perspective of this condition; whilst also proposing a future roadmap to delivering precision medicine-based management in ACM.
Collapse
Affiliation(s)
- Viraj Patel
- Department of Cardiology, Royal Papworth Hospital, Cambridge CB2 0AY, UK;
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
| | | | - Patricia B. Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK;
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Anjali Tiku-Owens
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.T.-O.); (P.S.); (D.M.); (F.E.M.)
| | - Luis R. Lopes
- Department of Cardiology, St Bartholomew’s Hospital, London EC1A 7BE, UK; (L.R.L.); (M.Y.K.); (A.P.)
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London WC1E 6BT, UK
| | - Mohammed Y. Khanji
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Department of Cardiology, St Bartholomew’s Hospital, London EC1A 7BE, UK; (L.R.L.); (M.Y.K.); (A.P.)
| | - Alexandros Protonotarios
- Department of Cardiology, St Bartholomew’s Hospital, London EC1A 7BE, UK; (L.R.L.); (M.Y.K.); (A.P.)
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London WC1E 6BT, UK
| | - Pasquale Santangeli
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.T.-O.); (P.S.); (D.M.); (F.E.M.)
| | - Daniele Muser
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.T.-O.); (P.S.); (D.M.); (F.E.M.)
| | - Francis E. Marchlinski
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.T.-O.); (P.S.); (D.M.); (F.E.M.)
| | - Peter A. Brady
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Division of Cardiology, Department of Medicine, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA
| | - C. Anwar A. Chahal
- Department of Cardiology, Royal Papworth Hospital, Cambridge CB2 0AY, UK;
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.T.-O.); (P.S.); (D.M.); (F.E.M.)
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Correspondence: ; Tel.: +1-267-252-3461
| |
Collapse
|
49
|
The Role of MicroRNAs in Arrhythmogenic Cardiomyopathy: Biomarkers or Innocent Bystanders of Disease Progression? Int J Mol Sci 2020; 21:ijms21176434. [PMID: 32899376 PMCID: PMC7504260 DOI: 10.3390/ijms21176434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an inherited cardiac disease characterized by a progressive fibro-fatty replacement of the working myocardium and by life-threatening arrhythmias and risk of sudden cardiac death. Pathogenic variants are identified in nearly 50% of affected patients mostly in genes encoding for desmosomal proteins. AC incomplete penetrance and phenotypic variability advocate that other factors than genetics may modulate the disease, such as microRNAs (miRNAs). MiRNAs are small noncoding RNAs with a primary role in gene expression regulation and network of cellular processes. The implication of miRNAs in AC pathogenesis and their role as biomarkers for early disease detection or differential diagnosis has been the objective of multiple studies employing diverse designs and methodologies to detect miRNAs and measure their expression levels. Here we summarize experiments, evidence, and flaws of the different studies and hitherto knowledge of the implication of miRNAs in AC pathogenesis and diagnosis.
Collapse
|
50
|
Ding Y, Yang J, Chen P, Lu T, Jiao K, Tester DJ, Giudicessi JR, Jiang K, Ackerman MJ, Li Y, Wang DW, Lee H, Wang DW, Xu X. Knockout of SORBS2 Protein Disrupts the Structural Integrity of Intercalated Disc and Manifests Features of Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2020; 9:e017055. [PMID: 32808564 PMCID: PMC7660791 DOI: 10.1161/jaha.119.017055] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Background Sorbs2b (sorbin and SH3 domain-containing 2b) was recently identified as a cardiomyopathy gene from a zebrafish mutagenesis screen. However, cardiac functions of its mammalian ortholog remain elusive. Methods and Results We conducted a detailed expression and subcellular localization analysis of Sorbs2 ortholog in mice and a phenotypic characterization in Sorbs2 knockout mice. Sorbs2 is highly expressed in the mouse heart and encodes an adhesion junction/desmosome protein that is mainly localized to the intercalated disc. A mutation with near complete depletion of the Sorbs2 protein in mice results in phenotypes characteristic of human arrhythmogenic cardiomyopathy (ACM), including right ventricular dilation, right ventricular dysfunction, spontaneous ventricular tachycardia, and premature death. Sorbs2 is required to maintain the structural integrity of intercalated disc. Its absence resulted in profound cardiac electrical remodeling with impaired impulse conduction and action potential derangements. Targeted sequencing of human patients with ACM identified 2 rare splicing variants classified as likely pathogenic were in 2 unrelated individuals with ACM from a cohort of 59 patients with ACM. Conclusions The Sorbs2 knockout mouse manifests several key features reminiscent of human ACM. Although the candidacy of SORBS2 as a new ACM-susceptibility gene is supported by preliminary human genetics study, future validation in larger cohorts with ACM is needed.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Jingchun Yang
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Peng Chen
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tong Lu
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Kunli Jiao
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | | | | | - Kai Jiang
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Michael J. Ackerman
- Department of Cardiovascular Medicine (Division of Heart Rhythm Services)Mayo ClinicRochesterMN
- Pediatric and Adolescent Medicine (Division of Pediatric Cardiology)Mayo ClinicRochesterMN
- Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory)Mayo ClinicRochesterMN
| | - Yigang Li
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Dao Wu Wang
- State Key Laboratory of Reproductive MedicineClinical Center of Reproductive Medicine and Department of CardiologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - HoN‐chi Lee
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Dao Wen Wang
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaolei Xu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| |
Collapse
|